1
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Cherepanov SA, Yusubalieva GM, Ruzsics Z, Lipatova AV, Chekhonin VP. Superior infectivity of the fiber chimeric oncolytic adenoviruses Ad5/35 and Ad5/3 over Ad5-delta-24-RGD in primary glioma cultures. Mol Ther Oncolytics 2022; 24:230-248. [PMID: 35071746 PMCID: PMC8761956 DOI: 10.1016/j.omto.2021.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/17/2021] [Indexed: 01/28/2023] Open
Abstract
Ad5-delta-24-RGD is currently the most clinically advanced recombinant adenovirus (rAd) for glioma therapy. We constructed a panel of fiber-modified rAds (Ad5RGD, Ad5/3, Ad5/35, Ad5/3RGD, and Ad5/35RGD, all harboring the delta-24 modification) and compared their infectivity, replication, reproduction, and cytolytic efficacy in human and rodent glioma cell lines and short-term cultures from primary gliomas. In human cells, both Ad5/35-delta-24 and Ad5/3-delta-24 displayed superior infectivity and cytolytic efficacy over Ad5-delta-24-RGD, while Ad5/3-delta-24-RGD and Ad5/35-delta-24-RGD did not show further improvements in efficacy. The expression of the adenoviral receptors/coreceptors CAR, DSG2, and CD46 and the integrins αVβ3/αVβ5 did not predict the relative cytolytic efficacy of the fiber-modified rAds. The cytotoxicity of the fiber-modified rAds in human primary normal cultures of different origins and in primary glioma cultures was comparable, indicating that the delta-24 modification did not confer tumor cell selectivity. We also revealed that CT-2A and GL261 glioma cells might be used as murine cell models for the fiber chimeric rAds in vitro and in vivo. In GL261 tumor-bearing mice, Ad5/35-delta-24, armed with the immune costimulator OX40L as the E2A/DBP-p2A-mOX40L fusion, produced long-term survivors, which were able to reject tumor cells upon rechallenge. Our data underscore the potential of local Ad5/35-delta-24-based immunovirotherapy for glioblastoma treatment.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov Str. 1, 117997 Moscow, Russia
- Corresponding author Aleksei A. Stepanenko, Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia.
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov Str. 1, 117997 Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia
| | - Sergey A. Cherepanov
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of Russia, Moscow, Russia
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov Str. 1, 117997 Moscow, Russia
| |
Collapse
|
2
|
Kloker LD, Yurttas C, Lauer UM. Three-dimensional tumor cell cultures employed in virotherapy research. Oncolytic Virother 2018; 7:79-93. [PMID: 30234074 PMCID: PMC6130269 DOI: 10.2147/ov.s165479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Oncolytic virotherapy constitutes an upcoming alternative treatment option for a broad spectrum of cancer entities. However, despite great research efforts, there is still only a single US Food and Drug Administration/European Medicines Agency-approved oncolytic virus available for clinical use. One reason for that is the gap between promising preclinical data and limited clinical success. Since oncolytic viruses are biological agents, they might require more realistic in vitro tumor models than common monolayer tumor cell cultures to provide meaningful predictive preclinical evaluation results. For more realistic invitro tumor models, three-dimensional tumor cell-culture systems can be employed in preclinical virotherapy research. This review provides an overview of spheroid and hydrogel tumor cell cultures, organotypic tumor-tissue slices, organotypic raft cultures, and tumor organoids utilized in the context of oncolytic virotherapy. Furthermore, we also discuss advantages, disadvantages, techniques, and difficulties of these three-dimensional tumor cell-culture systems when applied specifically in virotherapy research.
Collapse
Affiliation(s)
- Linus D Kloker
- Department of Clinical Tumor Biology, University Hospital, University of Tübingen, Tübingen, Germany,
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital, University of Tübingen, Tübingen, Germany
| | - Ulrich M Lauer
- Department of Clinical Tumor Biology, University Hospital, University of Tübingen, Tübingen, Germany, .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Tübingen, Germany,
| |
Collapse
|
3
|
Abstract
Oncolytic virus (OV) therapy utilizes replication-competent viruses to kill cancer cells, leaving non-malignant cells unharmed. With the first U.S. Food and Drug Administration-approved OV, dozens of clinical trials ongoing, and an abundance of translational research in the field, OV therapy is poised to be one of the leading treatments for cancer. A number of recombinant OVs expressing a transgene for p53 (TP53) or another p53 family member (TP63 or TP73) were engineered with the goal of generating more potent OVs that function synergistically with host immunity and/or other therapies to reduce or eliminate tumor burden. Such transgenes have proven effective at improving OV therapies, and basic research has shown mechanisms of p53-mediated enhancement of OV therapy, provided optimized p53 transgenes, explored drug-OV combinational treatments, and challenged canonical roles for p53 in virus-host interactions and tumor suppression. This review summarizes studies combining p53 gene therapy with replication-competent OV therapy, reviews preclinical and clinical studies with replication-deficient gene therapy vectors expressing p53 transgene, examines how wild-type p53 and p53 modifications affect OV replication and anti-tumor effects of OV therapy, and explores future directions for rational design of OV therapy combined with p53 gene therapy.
Collapse
|
4
|
Three-dimensional cell culture models for investigating human viruses. Virol Sin 2016; 31:363-379. [PMID: 27822716 PMCID: PMC7090760 DOI: 10.1007/s12250-016-3889-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
Three-dimensional (3D) culture models are physiologically relevant, as they provide reproducible results, experimental flexibility and can be adapted for high-throughput experiments. Moreover, these models bridge the gap between traditional two-dimensional (2D) monolayer cultures and animal models. 3D culture systems have significantly advanced basic cell science and tissue engineering, especially in the fields of cell biology and physiology, stem cell research, regenerative medicine, cancer research, drug discovery, and gene and protein expression studies. In addition, 3D models can provide unique insight into bacteriology, virology, parasitology and host-pathogen interactions. This review summarizes and analyzes recent progress in human virological research with 3D cell culture models. We discuss viral growth, replication, proliferation, infection, virus-host interactions and antiviral drugs in 3D culture models.
Collapse
|
5
|
LaRocca CJ, Han J, Salzwedel AO, Davydova J, Herzberg MC, Gopalakrishnan R, Yamamoto M. Oncolytic adenoviruses targeted to Human Papilloma Virus-positive head and neck squamous cell carcinomas. Oral Oncol 2016; 56:25-31. [PMID: 27086483 DOI: 10.1016/j.oraloncology.2016.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/18/2016] [Accepted: 02/29/2016] [Indexed: 01/31/2023]
Abstract
OBJECTIVES In recent years, the incidence of Human Papilloma Virus (HPV)-positive head and neck squamous cell carcinomas (HNSCC) has markedly increased. Our aim was to design a novel therapeutic agent through the use of conditionally replicative adenoviruses (CRAds) that are targeted to the HPV E6 and E7 oncoproteins. METHODS Each adenovirus included small deletion(s) in the E1a region of the genome (Δ24 or CB016) intended to allow for selective replication in HPV-positive cells. In vitro assays were performed to analyze the transduction efficiency of the vectors and the cell viability following viral infection. Then, the UPCI SCC090 cell line (HPV-positive) was used to establish subcutaneous tumors in the flanks of nude mice. The tumors were then treated with either one dose of the virus or four doses (injected every fourth day). RESULTS The transduction analysis with luciferase-expressing viruses demonstrated that the 5/3 fiber modification maximized virus infectivity. In vitro, both viruses (5/3Δ24 and 5/3CB016) demonstrated profound oncolytic effects. The 5/3CB016 virus was more selective for HPV-positive HNSCC cells, whereas the 5/3Δ24 virus killed HNSCC cells regardless of HPV status. In vivo, single injections of both viruses demonstrated anti-tumor effects for only a few days following viral inoculation. However, after four viral injections, there was statistically significant reductions in tumor growth when compared to the control group (p<0.05). CONCLUSION CRAds targeted to HPV-positive HNSCCs demonstrated excellent in vitro and in vivo therapeutic effects, and they have the potential to be clinically translated as a novel treatment modality for this emerging disease.
Collapse
Affiliation(s)
| | - Joohee Han
- Department of Surgery, University of Minnesota, United States
| | | | - Julia Davydova
- Department of Surgery, University of Minnesota, United States; Masonic Cancer Center, University of Minnesota, United States
| | - Mark C Herzberg
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, United States
| | - Rajaram Gopalakrishnan
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, United States
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, United States; Masonic Cancer Center, University of Minnesota, United States; Institute of Molecular Virology, University of Minnesota, United States.
| |
Collapse
|
6
|
Abstract
Oncolytic adenoviruses (Ads) constitute a promising new class of anticancer agent. They are based on the well-studied adenoviral vector system, which lends itself to concept-driven design to generate oncolytic variants. The first oncolytic Ad was approved as a drug in China in 2005, although clinical efficacy observed in human trials has failed to reach the high expectations that were based on studies in animal models. Current obstacles to the full realization of efficacy of this class of anticancer agent include (i) limited efficiency of infection and specific replication in tumor cells, (ii) limited vector spread within the tumor, (iii) imperfect animal models and methods of in vivo imaging, and (iv) an incomplete understanding of the interaction of these agents with the host. In this review, we discuss recent advances in the field of oncolytic Ads and potential ways to overcome current obstacles to their clinical application and efficacy.
Collapse
|
7
|
Song H, Moseley PL, Lowe SL, Ozbun MA. Inducible heat shock protein 70 enhances HPV31 viral genome replication and virion production during the differentiation-dependent life cycle in human keratinocytes. Virus Res 2009; 147:113-22. [PMID: 19896513 DOI: 10.1016/j.virusres.2009.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 10/22/2009] [Accepted: 10/22/2009] [Indexed: 01/31/2023]
Abstract
Increasing data indicate heat shock proteins (HSPs) including inducible HSP70 (HSP70i) are involved in the replicative cycles of various viruses including adenoviruses (Ads), polyomaviruses (PyVs), and some RNA viruses. Cell-free system studies implicate HSP70i in human papillomavirus type 11 (HPV11) genome replication with E1 and E2 proteins, and there is evidence that HSP70 is involved in capsid assembly and disassembly for PyVs and HPVs. HSP70 expression is increased in HPV16 E6/E7 gene transduced human primary keratinocytes, and frequently detected in early stage uterine cervical cancer at levels in conjunction with lesion severity. In this study we carry out analyses in the natural host epithelial tissues to assess the role of inducible HSP70 (HSP70i) in the HPV infectious life cycle. For these studies we used the organotypic (raft) culture system to recapitulate the full viral life cycle of the high-risk HPV31. Upon heat shock of HPV31-infected organotypic tissues, we find high and sustained expression of HSP70i coincident with enhanced HPV genome replication and virion production. Whereas there is no clear effect on L1 expression levels, we find HSP70i and L1 interact and HSP70i colocalizes with and enhances the nuclear localization of L1 in differentiated cells. Ad-mediated gene transfer was used to study the effects of HSP70i in naturally HPV-infected differentiating tissues and showed results similar to those in heat shocked rafts. These results indicate that increased HSP70i augments late activities in the viral life cycle. We conclude that HSP70i contributes directly to HPV replicative viral activities and the production of infectious virions.
Collapse
Affiliation(s)
- Hebin Song
- Department of Molecular Genetics and Microbiology, The University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | | | | | | |
Collapse
|
8
|
Epithelial raft cultures for investigations of virus growth, pathogenesis and efficacy of antiviral agents. Antiviral Res 2009; 85:431-49. [PMID: 19883696 DOI: 10.1016/j.antiviral.2009.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 10/20/2009] [Accepted: 10/22/2009] [Indexed: 12/12/2022]
Abstract
The organotypic epithelial raft cultures, originally developed to study keratinocytes differentiation, represent a novel approach to the study of viruses able to infect epithelial cells. Organotypic epithelial raft cultures accurately reproduce the process of epithelial differentiation in vitro and can be prepared from normal keratinocytes, explanted epithelial tissue, or established cell lines. This culture system permits cells to proliferate and fully differentiate at the air-liquid interface on a dermal-equivalent support. Normal primary human keratinocytes (PHKs) stratify and fully differentiate in a manner similar to the normal squamous epithelial tissues, while transformed cell lines exhibit dysplastic morphologies similar to the (pre)neoplastic lesions seen in vivo. This three-dimensional (3D) culture system provides an essential tool for investigations of virus growth, virus-host cell interactions, for the genetic analysis of viral proteins and regulatory sequences, and for the evaluation of antiviral agents. The 3D epithelial cultures have proven a breakthrough in the research on papillomaviruses, since their life cycle is strictly linked to the differentiation of the host epithelium. In the last years, several reports have shown the usefulness of the 3D epithelial cultures for the study of other viruses that target at least during a part of their life cycles epithelial cells. The 3D epithelial cultures allow the analysis of virus-host cell interactions in stratified epithelia that more closely resemble the in vivo situation. In this review we describe the advances on research on 3D epithelial cultures for the study of virus growth and pathogenesis of different families of viruses, including papilloma-, herpes-, pox-, adeno-, and parvoviruses.
Collapse
|
9
|
Dorer DE, Nettelbeck DM. Targeting cancer by transcriptional control in cancer gene therapy and viral oncolysis. Adv Drug Deliv Rev 2009; 61:554-71. [PMID: 19394376 DOI: 10.1016/j.addr.2009.03.013] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Accepted: 03/05/2009] [Indexed: 01/02/2023]
Abstract
Cancer-specificity is the key requirement for a drug or treatment regimen to be effective against malignant disease--and has rarely been achieved adequately to date. Therefore, targeting strategies need to be implemented for future therapies to ensure efficient activity at the site of patients' tumors or metastases without causing intolerable side-effects. Gene therapy and viral oncolysis represent treatment modalities that offer unique opportunities for tumor targeting. This is because both the transfer of genes with anti-cancer activity and viral replication-induced cell killing, respectively, facilitate the incorporation of multiple mechanisms restricting their activity to cancer. To this end, cellular mechanisms of gene regulation have been successfully exploited to direct therapeutic gene expression and viral cell lysis to cancer cells. Here, transcriptional targeting has been the role model and most widely investigated. This approach exploits cellular gene regulatory elements that mediate cell type-specific transcription to restrict the expression of therapeutic genes or essential viral genes, ideally to cancer cells. In this review, we first discuss the rationale for such promoter targeting and its limitations. We then give an overview how tissue-/tumor-specific promoters are being identified and characterized. Strategies to apply and optimize such promoters for the engineering of targeted viral gene transfer vectors and oncolytic viruses-with respect to promoter size, selectivity and activity in the context of viral genomes-are described. Finally, we discuss in more detail individual examples for transcriptionally targeted virus drugs. First highlighting oncolytic viruses targeted by prostate-specific promoters and by the telomerase promoter as representatives of tissue-targeted and pan-cancer-specific virus drugs respectively, and secondly recent developments of the last two years.
Collapse
Affiliation(s)
- Dominik E Dorer
- Helmholtz-University Group Oncolytic Adenoviruses, German Cancer Research Center (DKFZ) and Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | | |
Collapse
|
10
|
Gillet JP, Macadangdang B, Fathke RL, Gottesman MM, Kimchi-Sarfaty C. The development of gene therapy: from monogenic recessive disorders to complex diseases such as cancer. Methods Mol Biol 2009; 542:5-54. [PMID: 19565894 DOI: 10.1007/978-1-59745-561-9_1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
During the last 4 decades, gene therapy has moved from preclinical to clinical studies for many diseases ranging from monogenic recessive disorders such as hemophilia to more complex diseases such as cancer, cardiovascular disorders, and human immunodeficiency virus (HIV). To date, more than 1,340 gene therapy clinical trials have been completed, are ongoing, or have been approved in 28 countries, using more than 100 genes. Most of those clinical trials (66.5%) were aimed at the treatment of cancer. Early hype, failures, and tragic events have now largely been replaced by the necessary stepwise progress needed to realize clinical benefits. We now understand better the strengths and weaknesses of various gene transfer vectors; this facilitates the choice of appropriate vectors for individual diseases. Continuous advances in our understanding of tumor biology have allowed the development of elegant, more efficient, and less toxic treatment strategies. In this introductory chapter, we review the history of gene therapy since the early 1960s and present in detail two major recurring themes in gene therapy: (1) the development of vector and delivery systems and (2) the design of strategies to fight or cure particular diseases. The field of cancer gene therapy experienced an "awkward adolescence." Although this field has certainly not yet reached maturity, it still holds the potential of alleviating the suffering of many individuals with cancer.
Collapse
Affiliation(s)
- Jean-Pierre Gillet
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
11
|
Abstract
Gene therapy represents a potentially useful approach for the treatment of diseases refractory to conventional therapies. Various preclinical and clinical strategies have been explored for treatment of gynaecological diseases. Given the most severe unmet clinical need, much of the work has been performed with gynaecological cancers and ovarian cancer in particular. Although the safety of many treatment strategies has been demonstrated in early phase clinical trials, efficacy has been mostly limited heretofore. Major challenges include improving the vectors used with the aim of more effective and selective delivery. In addition, effective penetration into and spreading within advanced and complex tumour masses and metastases remains challenging. This review focuses on existing and developmental gene transfer applications for gynaecological diseases.
Collapse
Affiliation(s)
- Anna Kanerva
- University of Helsinki, Cancer Gene Therapy Group, Transplantation Laboratory and Haartman Institute, P.O. Box 63 (Haartmaninkatu 8, 00290 Helsinki), Biomedicum, Helsinki 00014, Finland
| | | | | |
Collapse
|
12
|
Carette JE, Graat HCA, Schagen FHE, Mastenbroek DCJ, Rots MG, Haisma HJ, Groothuis GMM, Schaap GR, Bras J, Kaspers GJL, Wuisman PIJM, Gerritsen WR, van Beusechem VW. A conditionally replicating adenovirus with strict selectivity in killing cells expressing epidermal growth factor receptor. Virology 2007; 361:56-67. [PMID: 17184803 DOI: 10.1016/j.virol.2006.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 09/20/2006] [Accepted: 11/10/2006] [Indexed: 11/24/2022]
Abstract
Virotherapy of cancer using oncolytic adenoviruses has shown promise in both preclinical and clinical settings. One important challenge to reach the full therapeutic potential of oncolytic adenoviruses is accomplishing efficient infection of cancer cells and avoiding uptake by normal tissue through tropism modification. Towards this goal, we constructed and characterized an oncolytic adenovirus, carrying mutated capsid proteins to abolish the promiscuous adenovirus native tropism and encoding a bispecific adapter molecule to target the virus to the epidermal growth factor receptor (EGFR). The new virus displayed a highly selective targeting profile, with reduced infection of EGFR-negative cells and efficient killing of EGFR-positive cancer cells including primary EGFR-positive osteosarcoma cells that are refractory to infection by conventional adenoviruses. Our method to modify adenovirus tropism might thus be useful to design new oncolytic adenoviruses for more effective treatment of cancer.
Collapse
Affiliation(s)
- Jan E Carette
- Department of Medical Oncology, Gene Therapy Division, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Delgado-Enciso I, Cervantes-García D, Martínez-Dávila IA, Ortiz-López R, Alemany-Bonastre R, Silva-Platas CI, Lugo-Trampe A, Barrera-Saldaña HA, Galván-Salazar HR, Coronel-Tene CG, Sánchez-Santillán CF, Rojas-Martínez A. A potent replicative delta-24 adenoviral vector driven by the promoter of human papillomavirus 16 that is highly selective for associated neoplasms. J Gene Med 2007; 9:852-61. [PMID: 17729237 DOI: 10.1002/jgm.1071] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Several human epithelial neoplasms are associated with high-risk strains of human papillomavirus (HPV) such as cervical, anorectal, and other carcinomas. For some tumor types the current therapeutic tools are only palliative. Conditionally replicative adenoviruses (CRAds) are promising antineoplastic agents, which also can trigger confined antitumor effects. METHODS We constructed a series of CRAds driven by the upstream regulatory promoter region (URR) of an Asian-American variant of HPV-16, which contained different mutations at the E1A region (dl1015 and/or Delta24) and wild-type. All vectors were tested in vitro for viral replication and cytotoxicity. Viral DNA replication and E1A expression were also assessed by quantitative PCR. Finally, we confirmed the antitumoral efficacy of this vector in injected and non-injected xenotransplanted cervical tumors in a murine model for tumor regression and survival studies. RESULTS A vector denominated Ad-URR/E1ADelta24 displayed a potent cytopathic effect associated with high selectivity for HPV+ cell lines. We found that the oncolytic effect of this CRAd was comparable to Ad-wt or Ad-Delta24, but this efficacy was significantly attenuated in HPV- cell lines, an effect that was contributed by the URR promoter. Ad-URR/E1ADelta24 was very effective to control tumor growth, in both, injected and non-injected tumors generated with two different HPV+ cell lines. CONCLUSIONS CRAd Ad-URR/E1ADelta24 is a highly selective vector for HPV+ cell lines and tumors that preserves the oncolytic efficacy of Ad-wt and Ad-Delta24. Our preclinical data suggest that this vector may be useful and safe for the treatment of tumors induced by HPV, like cervical cancers.
Collapse
Affiliation(s)
- Iván Delgado-Enciso
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Adenovirus (Ad)-based cancer gene therapy is a promising, novel approach for treating cancer resistant to established treatment modalities. Unfortunately, the efficacy of nonreplicative first generation Ads was low and data from clinical trials were disappointing. To address this problem, conditionally replicating Ads have been constructed. Infection of tumor cells with conditionally replicating Ads results in tumor-specific replication, subsequent oncolysis and release of the virus progeny. Recently, it has been suggested that the low expression of the coxsackie-Ad receptor is the rate-limiting factor for infectivity with serotype 5 (Ad5). Unfortunately, coxsackie-Ad receptor expression is highly variable and often low on many tumor types. Consequently, molecular strategies have been applied for the development of coxsackie-Ad receptor-independent oncolytic Ads. This review describes recent developments of Ad-based cancer gene therapy, including novel engineering techniques of the Ad capsid for efficient tumor targeting, as well as targeting techniques, to restrict transgene expression to cancer cells.
Collapse
Affiliation(s)
- Daniel T Rein
- University of Düsseldorf Medical Center, Department of Obstetrics and Gynecology, Düsseldorf, Germany
| | | | | |
Collapse
|
15
|
Liu HD, Zheng H, Li M, Hu DS, Tang M, Cao Y. Upregulated expression of kappa light chain by Epstein-Barr virus encoded latent membrane protein 1 in nasopharyngeal carcinoma cells via NF-kappaB and AP-1 pathways. Cell Signal 2006; 19:419-27. [PMID: 16979873 DOI: 10.1016/j.cellsig.2006.07.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Accepted: 07/24/2006] [Indexed: 12/13/2022]
Abstract
B lymphocytes are generally considered to be the only source of immunoglobulins. However, increasing evidence revealed that some human epithelial cancer cell lines, including nasopharyngeal carcinoma (NPC) cell lines, expressed immunoglobulins. Moreover, we previously found that expression of kappa light chain in NPC cells could be upregulated by EBV-encoded latent membrane protein 1 (LMP1). Here, Western blot and flow cytometric analysis of intracellular kappa staining indicated that upregulation of the expression of kappa was inhibited by using LMP1-targeted DNAzyme and that Bay11-7082 and SP600125, inhibitors of JNK and NF-kappaB, respectively, inhibited LMP1-augmented kappa light chain expression in NPC cells. LMP1-positive NPC cells expressing the dominant-negative mutant of IkappaBalpha (DNMIkappaBalpha) or of c-Jun (TAM67) exhibited significantly decreasing kappa production compared with their parental cells. These results suggest that LMP1 elevated kappa light chain through activation of the NF-kappaB and AP-1 signaling pathways. The present study provided some hints of possible mechanisms by which human cancer cells of epithelial origin produced immunoglobulins.
Collapse
Affiliation(s)
- Hai-dan Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road #110, Changsha, Hunan 410078, PR China
| | | | | | | | | | | |
Collapse
|
16
|
Andrei G. Three-dimensional culture models for human viral diseases and antiviral drug development. Antiviral Res 2006; 71:96-107. [PMID: 16844237 DOI: 10.1016/j.antiviral.2006.05.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 05/30/2006] [Accepted: 05/31/2006] [Indexed: 10/24/2022]
Abstract
Researchers are recognizing the limitations of two-dimensional (2D) cell cultures, given the fact that they do not reproduce the morphology and biochemical features that the cells possess in the original tissue. As an alternative, the three-dimensional (3D) cell culture approach offers researchers the possibility to study cell growth and differentiation under conditions that more closely resemble the in vivo situation with regard to cell shape and cellular environment. Currently, 3D culture models are being employed in many areas of biomedical research because they offer a more realistic milieu than 2D cultures. The era of 2D culture techniques is moving towards a new epoch of culture systems in 3D. The present review is focused on topics of research on 3D cell cultures in virology and their use in antiviral drug development.
Collapse
|
17
|
Raki M, Rein DT, Kanerva A, Hemminki A. Gene Transfer Approaches for Gynecological Diseases. Mol Ther 2006; 14:154-63. [PMID: 16650808 DOI: 10.1016/j.ymthe.2006.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 12/13/2005] [Accepted: 02/06/2006] [Indexed: 11/25/2022] Open
Abstract
Gene transfer presents a potentially useful approach for the treatment of diseases refractory to conventional therapies. Various preclinical and clinical strategies have been explored for treatment of gynecological diseases. Given the direst need for novel treatments, much of the work has been performed with gynecological cancers and ovarian cancer in particular. Although the safety of many approaches has been demonstrated in early phase clinical trials, efficacy has been mostly limited so far. Major challenges include improving gene transfer vectors for enhanced and selective delivery and achieving effective penetration and spread within advanced and complex tumor masses. This review will focus on current and developmental gene transfer applications for gynecological diseases.
Collapse
Affiliation(s)
- Mari Raki
- Cancer Gene Therapy Group, Rational Drug Design Program, University of Helsinki, 00014 Helsinki, Finland
| | | | | | | |
Collapse
|
18
|
Bauerschmitz GJ, Guse K, Kanerva A, Menzel A, Herrmann I, Desmond RA, Yamamoto M, Nettelbeck DM, Hakkarainen T, Dall P, Curiel DT, Hemminki A. Triple-Targeted Oncolytic Adenoviruses Featuring the Cox2 Promoter, E1A Transcomplementation, and Serotype Chimerism for Enhanced Selectivity for Ovarian Cancer Cells. Mol Ther 2006; 14:164-74. [PMID: 16580264 DOI: 10.1016/j.ymthe.2006.01.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Revised: 01/12/2006] [Accepted: 01/30/2006] [Indexed: 01/24/2023] Open
Abstract
Conditionally replicating adenoviruses (CRAd's) feature selective replication in and killing of tumor cells. Initial clinical studies with relatively attenuated early generation agents have resulted in promising safety and efficacy data. Nevertheless, increased specificity may be advantageous for an emerging generation of infectivity-enhanced CRAd's. Further, increased specificity could translate into a larger tolerated dose. An approach for increasing specificity is dual control of E1A expression. We constructed six CRAd's featuring two variants of the cyclo-oxygenase 2 (cox2) promoter, combined with three versions of E1A. Transcriptional targeting was supplemented with transductional targeting utilizing the serotype 3 knob. In vivo and in vitro results suggest that cox2 can be utilized for enhancing the specificity of E1A deletion mutants and that combination with the Delta24 mutation increases specificity without reducing potency. Combination with Delta2-Delta24 was specific but somewhat attenuated. The promoter variants behaved similarly, although the longer 1,554-bp version displayed a trend for improved specificity. Transcriptional modifications were compatible with transductional targeting and resulted in up to 100,000-fold increase in the therapeutic window for Ad5/3cox2Ld24 vs wild-type adenovirus. Thus, the proposed triple-targeting strategy may be useful for increasing the safety and efficacy of adenoviral gene therapy for ovarian cancer.
Collapse
Affiliation(s)
- Gerd J Bauerschmitz
- Department of Medicine, Division of Human Gene Therapy, Gene Therapy Center, University of Alabama at Birmingham, 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Majem M, Cascallo M, Bayo-Puxan N, Mesia R, Germa JR, Alemany R. Control of E1A under an E2F-1 promoter insulated with the myotonic dystrophy locus insulator reduces the toxicity of oncolytic adenovirus Ad-Δ24RGD. Cancer Gene Ther 2006; 13:696-705. [PMID: 16498429 DOI: 10.1038/sj.cgt.7700940] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We previously described Ad-Delta24RGD as an enhanced-infectivity oncolytic adenovirus that targets tumors with an impaired RB pathway. The common alteration of this pathway in cancer eliminates the interaction of pRB with E2F and releases free E2F to activate E2F-responsive promoters, including the E2F-1 promoter. To improve the selectivity towards RB pathway-defective tumors and reduce the toxicity of Ad-Delta24RGD we aimed to control E1A-Delta24 expression under the E2F-1 promoter. A polyA signal was inserted upstream of the E2F-1 promoter to stop transcription initiated at the adenovirus ITR and packaging signal. The human myotonic dystropy locus insulator (DM-1) was also located between the E1a enhancers and the E2F-1 promoter to further insulate the promoter. The Ad-Delta24RGD derivative containing these insulation sequences expressed less E1a-Delta24 in normal cells and resulted less toxic while maintaining the potent oncolytic activity of the parental virus. These results demonstrate that the human DM-1 inslulator can function in an adenovirus context to maintain heterologous promoter selectivity. The new oncolytic adenovirus presented here may represent a valuable therapeutic option for a broad range of tumors with a deregulated E2F/pRB pathway.
Collapse
Affiliation(s)
- M Majem
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
20
|
Shimony N, Gorodetsky R, Marx G, Gal D, Rivkin R, Ben-Ari A, Landsman A, Haviv YS. Fibrin microbeads (FMB) as a 3D platform for kidney gene and cell therapy. Kidney Int 2006; 69:625-33. [PMID: 16395256 DOI: 10.1038/sj.ki.5000099] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cell and gene therapy may alter the outcome of renal diseases, such as hereditary nephropathies, acute and chronic glomerulonephritis and allograft nephropathy. However, owing to blockade of many viral and cellular vehicles by the complex glomerular architecture, the exact nature of gene and cell delivery into specific renal compartments remains currently unknown. To study the interaction of viral vectors with a variety of renal cells and mesenchymal stem cells (MSCs), we employed a novel biological three-dimensional (3D) matrix comprised of fibrin microbeads (FMB) in comparison to monolayer cell culture. Our studies showed that renal cells of both established and primary lines can grow efficiently on FMB and differentiate into epithelial structures, as shown by electron microscopy. Gene delivery into renal cells in 3D was observed for several viral vectors and growth in 3D on FMB conferred resistance to renal cancer cells in the context of oncolytic adenoviruses. Finally, MSCs from various rodent species attached to FMB, grew robustly, survived for several weeks and could efficiently be transduced on FMB. Thus, on the basis of growth, differentiation and transduction of renal cells in 3D, FMB emerge as a novel 3D cellular microenvironment that differs substantially from monolayer cell cultures.
Collapse
Affiliation(s)
- N Shimony
- Cell and Gene Therapy Program, Department of Medicine, Division of Nephrology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Stoff-Khalili MA, Rivera AA, Le LP, Stoff A, Everts M, Contreras JL, Chen D, Teng L, Rots MG, Haisma HJ, Rocconi RP, Bauerschmitz GJ, Rein DT, Yamamoto M, Siegal GP, Dall P, Michael Mathis J, Curiel DT. Employment of liver tissue slice analysis to assay hepatotoxicity linked to replicative and nonreplicative adenoviral agents. Cancer Gene Ther 2006; 13:606-18. [PMID: 16410819 DOI: 10.1038/sj.cgt.7700934] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Whereas virotherapy has emerged as a novel and promising approach for neoplastic diseases, appropriate model systems have hampered preclinical evaluation of candidate conditionally replicative adenovirus agents (CRAds) with respect to liver toxicity. This is due to the inability of human viral agents to cross species. We have recently shown the human liver tissue slice model to be a facile means to validate adenoviral replication. On this basis, we sought to determine whether our ex vivo liver tissue slice model could be used to assess CRAd-mediated liver toxicity. We analyzed and compared the toxicity of a conditionally replicative adenovirus (AdDelta24) to that of a replication incompetent adenovirus (Adnull [E1-]) in mouse and human liver tissue slices. To accomplish this, we examined the hepatic apoptosis expression profile by DNA microarray analyses, and compared these results to extracellular release of aminotransferase enzymes, along with direct evidence of apoptosis by caspase-3 immunhistochemical staining and TUNEL assays. Human and mouse liver tissue slices demonstrated a marked increase in extracellular release of aminotransferase enzymes on infection with AdDelta24 compared to Adnull. AdDelta24-mediated liver toxicity was further demonstrated by apoptosis induction, as detected by caspase-3 immunohistochemical staining, TUNEL assay and microarray analysis. In conclusion, concordance of CRAd-mediated apoptosis in both the human and the mouse liver tissue slice models was demonstrated, despite the limited replication ability of CRAds in mouse liver slices. The results of this study, defining the CRAd-mediated apoptosis gene expression profiles in human and mouse liver, may lay a foundation for preclinical liver toxicity analysis of CRAd agents.
Collapse
Affiliation(s)
- M A Stoff-Khalili
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA, and Department of Plastic and Reconstructive Surgery, Dreifaltigkeits-Hospital, Wesseling, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The application of replicating viruses for the treatment of cancers represents a novel therapy that is distinct from traditional treatment modalities. It is apparent that the genetic changes that a virus produces within an infected cell in order to create an environment conducive to viral replication are often similar to the processes involved in cellular transformation. These include uncontrolled cellular proliferation, prevention of apoptosis, and resistance to host organism immune effector mechanisms. Deletions of viral genes involved in these processes have been exploited to produce viral mutants whose replication is selective for transformed cells. The use of tissue-specific transcriptional response or RNA stability elements to control the expression of critical viral genes has also resulted in targeted viruses. Work also is being undertaken to restrict or alter the tropism of viruses by altering their ability to infect certain cell types. Finally, the addition of exogenous genes can be used to increase the virus's lytic potential and/or bystander killing; to further induce the host's immune response against cancer cells; and/or to permit the controlled downregulation of viral replication if necessary. The combination of different tumor-targeting mutations in parallel with the expression of foreign genes has resulted in the evolution of second- and third-generation viruses that continue to become further distinct from their native parental strains. The movement of these viruses into the clinic has begun to demonstrate the potential of this approach in the treatment of cancers.
Collapse
Affiliation(s)
- Stephen H Thorne
- Department of Pediatrics and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305-5427, USA.
| | | | | |
Collapse
|
23
|
Jiang H, Alemany R, Gomez-Manzano C, Medrano DR, Lemoine MG, Olson MV, Alonso MM, Lee OH, Conrad CC, Yung WKA, Fueyo J. Downmodulation of E1A protein expression as a novel strategy to design cancer-selective adenoviruses. Neoplasia 2005; 7:723-9. [PMID: 16207474 PMCID: PMC1501891 DOI: 10.1593/neo.04793] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Accepted: 03/11/2005] [Indexed: 12/18/2022] Open
Abstract
Oncolytic adenoviruses are being tested as potential therapies for human malignant tumors, including gliomas. Here we report for the first time that a mutation in the E1A gene results in low levels of E1A protein, conditioning the replication of mutant adenoviruses specifically to cancer cells. In this study, we compared the oncolytic potencies of three mutant adenoviruses encompassing deletions within the CR1 (Delta-39), CR2 (Delta-24) regions, or both regions (Delta-24/39) of the E1A protein. Delta-39 and Delta-24 induced a cytopathic effect with similar efficiency in glioma cells and a comparable capacity for replication. Importantly, the activity of Delta-39 was significantly attenuated compared to Delta-24 in proliferating normal human astrocytes. Direct analyses of the activation of E2F-1 promoter demonstrated the inability of Delta-39 to induce S-phase-related transcriptional activity in normal cells. Interestingly, E1A protein levels in cells infected with Delta-39 were remarkably downmodulated. Furthermore, protein stability studies revealed enhanced degradation of CR1 mutant E1A proteins, and inhibition of the proteasome activity resulted in the striking rescue of E1A levels. We conclude that the level of E1A protein is a critical determinant of oncolytic phenotype and we propose a completely novel strategy for the design and construction of conditionally replicative adenoviruses.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Diana R Medrano
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Michael G Lemoine
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Melissa V Olson
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Marta M Alonso
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Ok-Hee Lee
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Charles C Conrad
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - W K Alfred Yung
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, Brain Tumor Center, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Abstract
The human papillomaviruses (HPVs) are a diverse group of infectious agents, some of which are a causative agent of human cancers. Cervical cancer and oral cancer are closely associated with specific types of HPV, and the tumors grow only if there is continual expression of the viral E6 and E7 genes. Evidence from in vitro studies shows that when expression of these genes is inhibited by gene therapy approaches such as antisense RNA, ribozymes, or siRNA, the transformed phenotype of the cells is lost. Although it seems possible that clinical applications of this approach could help in the management of cervical and oral cancers there have been no clinical trials of gene therapy for HPV-associated cancers. Since the basic information is now available, a shift to translational research would be greatly welcomed.
Collapse
Affiliation(s)
- E J Shillitoe
- Department of Microbiology & Immunology, Upstate Medical University, State University of New York, Syracuse, NY 13210, USA.
| |
Collapse
|
25
|
Heideman DAM, Steenbergen RDM, van der Torre J, Scheffner M, Alemany R, Gerritsen WR, Meijer CJLM, Snijders PJF, van Beusechem VW. Oncolytic Adenovirus Expressing a p53 Variant Resistant to Degradation by HPV E6 Protein Exhibits Potent and Selective Replication in Cervical Cancer. Mol Ther 2005; 12:1083-90. [PMID: 16085463 DOI: 10.1016/j.ymthe.2005.06.443] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 06/06/2005] [Accepted: 06/07/2005] [Indexed: 10/25/2022] Open
Abstract
Rationale in the development of novel treatment strategies for HPV-associated cancers is targeting on the basis of the presence of HPV in (pre)malignant cells. Here, we designed a new conditionally replicating adenovirus (CRAd) for selective and effective oncolytic replication in HPV-containing cells. As the backbone, we used the CRAd AdCB016, which replicates selectively in cells expressing HPV E6 and E7 proteins. To enhance its oncolytic potency, we armed AdCB016 with p53 variant mp53(268N), which is resistant to HPV E6-mediated degradation. The new CRAd AdCB016-mp53(268N) was analyzed for its lytic replication properties in cervical carcinoma cell lines, HPV-immortalized keratinocyte cell lines representing dysplastic cells, and primary human keratinocytes. AdCB016-mp53(268N) exhibited 10- to 1000-fold greater efficacy than AdCB016 on high-risk HPV-positive cervical carcinoma cells and HPV-immortalized keratinocytes. Importantly, infection with AdCB016-mp53(268N) did not affect primary nonmalignant human keratinocytes. This favorable efficacy and safety profile was confirmed in organotypic raft cultures. Our findings suggest that AdCB016-mp53(268N) is a promising new agent for treatment of HPV-associated human cancers.
Collapse
Affiliation(s)
- Daniëlle A M Heideman
- Department of Pathology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhou J, Gao Q, Chen G, Huang X, Lu Y, Li K, Xie D, Zhuang L, Deng J, Ma D. Novel oncolytic adenovirus selectively targets tumor-associated polo-like kinase 1 and tumor cell viability. Clin Cancer Res 2005; 11:8431-40. [PMID: 16322306 DOI: 10.1158/1078-0432.ccr-05-1085] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Polo-like kinase 1 (plk1) is a serine/threonine protein kinase essential for multiple mitotic processes. Previous observations have validated plk1 as a promising therapeutic target. Despite being conceptually attractive, the potency and specificity of current plk1-based therapies remain limited. We sought to develop a novel plk1-targeting strategy by constructing an oncolytic adenovirus to selectively silence plk1 in tumor cells. EXPERIMENTAL DESIGN Two artificial features were engineered into one wild-type adenovirus type 5 (wt-Adv5) genome to generate a new oncolytic adenovirus (M1). First, M1 contains a 27-bp deletion in E1A region, which confers potent, oncolytic efficacy. Second, M1 is armed with a fragment of antisense plk1 cDNA that substitutes the E3 region encoding 6.7K and gp19K. In this design, tumor-selective replication of M1 would activate the native adenovirus E3 promoters to express the antisense plk1 cDNA preferentially in tumor cells and silence tumor-associated plk1 protein. RESULTS By virtue of combining oncolysis with plk1 targeting, M1 exhibited potent antitumoral efficacy in vitro and in vivo. Systemic administration of M1 plus cisplatin induced complete tumor regression in 80% of orthotopic hepatic carcinoma model mice that were otherwise resistant to cisplatin and disseminated metastases. CONCLUSIONS Coupling plk1 targeting with oncolysis had shown superior antitumor efficacy. Present findings would benefit the development of novel oncolytic adenoviruses generally applicable to a wide range of molecule-based therapeutics.
Collapse
Affiliation(s)
- Jianfeng Zhou
- Cancer Biology Research Center, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, WuHan, Hubei, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Howe JA, Pelka P, Antelman D, Wilson C, Cornell D, Hancock W, Ramachandra M, Avanzini J, Horn M, Wills K, Sutjipto S, Ralston R. Matching complementing functions of transformed cells with stable expression of selected viral genes for production of E1-deleted adenovirus vectors. Virology 2005; 345:220-30. [PMID: 16246392 DOI: 10.1016/j.virol.2005.09.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 08/23/2005] [Accepted: 09/08/2005] [Indexed: 10/25/2022]
Abstract
Production of E1-deleted adenovirus (rAd) vectors requires complementation by E1A and E1B functions provided by the production cell line. The two cell lines most commonly used for production of rAd vectors, 293 and Per.C6, were derived from human primary cells and contain contiguous E1A and E1B sequences from the Ad genome. As an alternative system, we tested complementation of rAd vectors using sequential transfection of individual E1A and E1B expression cassettes into A549 human lung tumor cells, which support highly efficient replication of wild type adenovirus. We found that E1A function could be complemented in A549 cells by the mutant E1Adl01/07, and that E1B function could be provided in such cells using only the 55K E1B gene. Production yields in the resulting producer cell line, designated SL0003, were similar to those obtained from 293 cells without generation of detectable recombinant replication competent adenovirus.
Collapse
Affiliation(s)
- John A Howe
- Biological Research, Virology, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The prognosis of patients with some kinds of cancers whose patients are often found unresectable upon diagnosis is still dismal. In these fields, development of a new therapeutic modality is needed and gene therapy represents one promising strategy. So far, numerous cancer gene therapy clinical trials based on these principles have been carried out and have shown the safety of such modalities, but have fallen short of the initial expectations to cure cancers. In this review, we would like to make a problem-oriented discussion of current status of cancer gene therapy research by using mainly gastrointestinal cancers as an example. In order to overcome obstacles for full realization of cancer gene therapy, numerous researches have been conducted by many researchers. Various cancer-selective and non-selective genes, as well as lytic viruses themselves have been employed for gene therapy. In the context of gene delivery method, different kinds of viral and non-viral strategies have been utilized. In addition, surrogate assays, such as soluble markers and imaging, have been developed for safer and more informative clinical trials. Many experiments and clinical trials to date have figured out current obstacles for the realization of an effective cancer gene therapy modality. Tireless efforts to overcome such hurdles and continuous infusion of novel concepts into this field should lead to break through technologies and the cure of the patients.
Collapse
Affiliation(s)
- Masato Yamamoto
- BMR2-410, 901 19th Street South, Birmingham, AL 35294-2172, USA
| | | |
Collapse
|
29
|
Abstract
The clinical outcome of advanced gastrointestinal (GI) cancers (especially pancreatic and oesophageal cancers) is dismal, despite the advance of conventional therapeutic strategies. Cancer gene therapy is a category of new therapeutics, among which conditionally replicative adenovirus (CRAd) is one promising strategy to overcome existing obstacles of cancer gene therapy. Various CRAds have been developed for GI cancer treatment by taking advantage of the replication biology of adenovirus. Some CRAds have already been tested in clinical trials, but have fallen short of initial expectations. Concerns for clinical applicability include therapeutic potency, replication selectivity and interval end points in clinical trials. In addition, improvement of experimental animal models is needed for a deeper understanding of CRAd biology. Despite these obstacles, CRAds continue to be an exciting area of investigation with great potential for clinical utility. Further virological and oncological research will eventually lead to full realisation of the therapeutic potential of CRAds in the field of GI cancers.
Collapse
Affiliation(s)
- Masato Yamamoto
- Division of Human Gene Therapy, Department of Medicine, and the Gene Therapy Center, University of Alabama at Birmingham, BMR2-408, 901 19th Street South, Birmingham, AL 35294-2172, USA.
| |
Collapse
|
30
|
Oosterhoff D, Pinedo HM, Witlox MA, Carette JE, Gerritsen WR, van Beusechem VW. Gene-directed enzyme prodrug therapy with carboxylesterase enhances the anticancer efficacy of the conditionally replicating adenovirus AdΔ24. Gene Ther 2005; 12:1011-8. [PMID: 15729367 DOI: 10.1038/sj.gt.3302492] [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: 11/09/2022]
Abstract
Conditionally replicating adenoviruses (CRAds) selectively replicate in and thereby kill cancer cells. The CRAd AdDelta24 with pRb-binding-deficient E1A kills cancer cells efficiently. Arming CRAds with genes encoding prodrug-converting enzymes could allow for enhanced anticancer efficacy by the combined effects of oncolytic replication and local prodrug activation. Here, we investigated combination treatment of human colon cancer cell lines with AdDelta24-type CRAds and gene-directed enzyme prodrug therapy (GDEPT) using two different enzyme/prodrug systems, that is, thymidine kinase/ganciclovir (TK/GCV) and carboxylesterase (CE)/CPT-11. On all three cell lines tested, GDEPT with TK/GCV made CRAd treatment less efficacious. In contrast, expression of a secreted form of CE (sCE2) combined with CPT-11 treatment markedly enhanced the efficacy of AdDelta24 virotherapy. Based on this observation, we constructed an AdDelta24 variant expressing sCE2. In the absence of CPT-11, this new CRAd Ad5-Delta24.E3-sCE2 was similarly effective as its parent in killing human colon cancer cells. Low concentrations of CPT-11 inhibited Ad5-Delta24.E3-sCE2 propagation. Nevertheless, CPT-11 specifically augmented the cytotoxicity of Ad5-Delta24.E3-sCE2 against all three-colon cancer cell lines. Hence, the positive contribution of sCE2/CPT-11 GDEPT to colon cancer cytotoxicity outweighed its negative influence on CRAd propagation. Therefore, CRAd-sCE2/CPT-11 combination therapy appears useful for more effective treatment of colon cancer.
Collapse
Affiliation(s)
- D Oosterhoff
- Division of Gene Therapy, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
31
|
Sauthoff H, Pipiya T, Heitner S, Chen S, Bleck B, Reibman J, Chang W, Norman RG, Rom WN, Hay JG. Impact of E1a Modifications on Tumor-Selective Adenoviral Replication and Toxicity. Mol Ther 2004; 10:749-57. [PMID: 15451459 DOI: 10.1016/j.ymthe.2004.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2003] [Accepted: 07/13/2004] [Indexed: 11/18/2022] Open
Abstract
Replicating adenoviral vectors are capable of multiplying up to a thousandfold in the target cell, a property that might prove to be of tremendous potential for cancer therapy. However, restricting viral replication and toxicity to cancer cells is essential to optimize safety. It has been proposed that modifications of the E1a protein that impair binding to Rb or p300 will prevent S-phase induction in normal cells, resulting in selective viral replication in tumor cells. However, it remains uncertain which of the several possible E1a modifications would be most effective at protecting normal cells without compromising the oncolytic effect of the vector. In this study, we have expressed several E1a-deletion mutants at high levels using the CMV promoter and tested them for their ability to facilitate S-phase induction, viral replication, and cytotoxicity in both normal and cancer cells. Deletion of the Rb-binding domain within E1a only slightly decreased the ability of the virus to induce S phase in growth-arrested cells. The effect of this deletion on viral replication and cytotoxicity was variable. There was reduced cytotoxicity in normal bronchial epithelial cells; however, in some normal cell types there was equal viral replication and cytotoxicity compared with wild type. Deletions in both the N-terminus and the Rb-binding domain were required to block S-phase induction effectively in growth-arrested normal cells; in addition, this virus demonstrated reduced viral replication and cytotoxicity in normal cells. An equally favorable replication and cytotoxicity profile was induced by a virus expressing E1a that is incapable of binding to the transcriptional adapter motif (TRAM) of p300. All viruses were equally cytotoxic to cancer cells compared with wild-type virus. In conclusion, deletion of the Rb-binding site alone within E1a may not be the most efficacious means of targeting viral replication and toxicity. However, deletion within the N-terminus in conjunction with a deletion within the Rb-binding domain, or deletion of the p300-TRAM binding domain, induces a more favorable cytotoxicity profile.
Collapse
Affiliation(s)
- Harald Sauthoff
- Division of Pulmonary & Critical Care Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Toth K, Djeha H, Ying B, Tollefson AE, Kuppuswamy M, Doronin K, Krajcsi P, Lipinski K, Wrighton CJ, Wold WSM. An oncolytic adenovirus vector combining enhanced cell-to-cell spreading, mediated by the ADP cytolytic protein, with selective replication in cancer cells with deregulated wnt signaling. Cancer Res 2004; 64:3638-44. [PMID: 15150123 DOI: 10.1158/0008-5472.can-03-3882] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have constructed a novel oncolytic adenovirus (Ad) vector named VRX-009 that combines enhanced cell spread with tumor-specific replication. Enhanced spread, which could significantly increase antitumor efficacy, is mediated by overexpression of the Ad cytolytic protein named ADP (also known as E3-11.6K). Replication of VRX-009 is restricted to cells with a deregulated wnt signal transduction pathway by replacement of the wild-type Ad E4 promoter with a synthetic promoter consisting of five consensus binding sites for the T-cell factor transcription factor. Tumor-selective replication is indicated by several lines of evidence. VRX-009 expresses E4ORF3, a representative Ad E4 protein, only in colon cancer cell lines. Furthermore, VRX-009 replicates preferentially in colon cancer cell lines as evidenced by virus productivity 2 orders of magnitude higher in SW480 colon cancer cells than in A549 lung cancer cells. Replication in primary human bronchial epithelial cells and human umbilical vein endothelial cells was also significantly lower than in SW480 cells. When tested in human tumor xenografts in nude mice, VRX-009 effectively suppressed the growth of SW480 colon tumors but not of A549 lung tumors. VRX-009 may provide greater level of antitumor efficacy than standard oncolytic Ad vectors in tumors in which a defect in wnt signaling increases the level of nuclear beta-catenin.
Collapse
Affiliation(s)
- Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri 63126, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gomez-Manzano C, Balague C, Alemany R, Lemoine MG, Mitlianga P, Jiang H, Khan A, Alonso M, Lang FF, Conrad CA, Liu TJ, Bekele BN, Yung WKA, Fueyo J. A novel E1A-E1B mutant adenovirus induces glioma regression in vivo. Oncogene 2004; 23:1821-8. [PMID: 15014451 DOI: 10.1038/sj.onc.1207321] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malignant gliomas are the most frequently occurring primary brain tumors and are resistant to conventional therapy. Conditionally replicating adenoviruses are a novel strategy in glioma treatment. Clinical trials using E1B mutant adenoviruses have been reported recently and E1A mutant replication-competent adenoviruses are in advanced preclinical testing. Here we constructed a novel replication-selective adenovirus (CB1) incorporating a double deletion of a 24 bp Rb-binding region in the E1a gene, and a 903 bp deleted region in the E1b gene that abrogates the expression of a p53-binding E1B-55 kDa protein. CB1 exerted a potent anticancer effect in vitro in U-251 MG, U-373 MG, and D-54 MG human glioma cell lines, as assessed by qualitative and quantitative viability assays. Replication analyses demonstrated that CB1 replicates in vitro in human glioma cells. Importantly, CB1 acquired a highly attenuated replicative phenotype in both serum-starved and proliferating normal human astrocytes. In vivo experiments using intracranially implanted D-54 MG glioma xenografts in nude mice showed that a single dose of CB1 (1.5 x 10(8) PFU/tumor) significantly improved survival. Immunohistochemical analyses of expressed adenoviral proteins confirmed adenoviral replication within the tumors. The CB1 oncolytic adenovirus induces a potent antiglioma effect and could ultimately demonstrate clinical relevance and therapeutic utility.
Collapse
Affiliation(s)
- Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Banerjee NS, Rivera AA, Wang M, Chow LT, Broker TR, Curiel DT, Nettelbeck DM. Analyses of melanoma-targeted oncolytic adenoviruses with tyrosinase enhancer/promoter-driven E1A, E4, or both in submerged cells and organotypic cultures. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.437.3.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
We have generated novel conditionally replicative adenoviruses (CRAds) targeted to melanoma cells. In these adenoviruses, the E4 region (AdΔ24TyrE4) or both E1 and E4 regions (Ad2xTyr) were controlled by a synthetic tyrosinase enhancer/promoter (Tyr2E/P) specific for melanocytes. The properties of these CRAds were compared with wild-type adenovirus (Adwt) and our previous CRAd with a targeted E1A CRII mutation (AdTyrΔ24) in submerged cultures of melanoma cells and nonmelanoma control cells. We showed that AdΔ24TyrE4 had a cell type selectivity similar to AdTyrΔ24 but had a distinct block in viral reproduction in nonmelanoma cells and that Ad2xTyr had an augmented selectivity for melanoma cells. These viruses were additionally tested in organotypic cultures of melanoma cell lines, primary human keratinocytes (PHKs), or mixed cell populations. Unexpectedly, the CRAds exhibited somewhat different cell type selectivity profiles in these cultures relative to those observed in submerged cultures, demonstrating the importance of multiple assay systems. Specifically, AdTyrΔ24 and Ad2xTyr were selective for melanoma cells, whereas AdΔ24TyrE4 exhibited no selectivity, similar to Adwt. AdTyrΔ24 and Ad2xTyr were strongly attenuated in their ability to lyse PHKs in organotypic cultures. Furthermore, Ad2xTyr had a superior melanoma selectivity in organotypic cultures of cocultivated melanoma cells and PHKs. The enhanced selectivity for melanoma cells exhibited by Ad2xTyr provides a window of opportunity for therapeutic application. These studies also demonstrate that organotypic cultures derived from mixtures of tumor and normal cells represent a promising new model for analysis of CRAd specificity and toxicity.
Collapse
Affiliation(s)
| | - Angel A. Rivera
- 2Division of Human Gene Therapy, Departments of Medicine, Pathology, and Surgery, and Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL and
| | - Minghui Wang
- 2Division of Human Gene Therapy, Departments of Medicine, Pathology, and Surgery, and Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL and
| | | | | | - David T. Curiel
- 2Division of Human Gene Therapy, Departments of Medicine, Pathology, and Surgery, and Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL and
| | - Dirk M. Nettelbeck
- 2Division of Human Gene Therapy, Departments of Medicine, Pathology, and Surgery, and Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL and
- 3Department of Dermatology, University Medical Center Erlangen, Erlangen, Germany
| |
Collapse
|
35
|
Abstract
Viral oncolysis, or virotherapy, is an endeavor to use viruses as therapeutic agents in an effort to exploit their highly evolved qualities of host cell killing and simultaneous multiplication and spread. This review describes the concept of oncolytic adenoviruses, also called conditionally replicative adenoviruses (CRAds), and recent developments--inspired by early clinical results--that aim at the optimization of CRAd efficacy. Molecular strategies applied for the development of oncolytic adenoviruses include (i) the genetic manipulation of the expression and/or function of key regulatory viral proteins in order to restrict viral replication and spread to tumor cells, (ii) the engineering of the adenoviral capsid for efficient and tumor-targeted infection, and (iii) the incorporation of heterologous genes to facilitate combination therapies or tracking of the virus. Initial clinical trials have provided proof-of-concept for adenoviral oncolysis in patients and a favorable safety profile for oncolytic adenoviruses has been demonstrated. In conclusion, adenoviral oncolysis, with its distinct therapeutic mechanism, shows remarkable therapeutic potential. Advanced generations of virotherapeutics are currently in development.
Collapse
Affiliation(s)
- Dirk M Nettelbeck
- Department of Dermatology, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
36
|
Vecil GG, Lang FF. Clinical trials of adenoviruses in brain tumors: a review of Ad-p53 and oncolytic adenoviruses. J Neurooncol 2004; 65:237-46. [PMID: 14682374 DOI: 10.1023/b:neon.0000003653.45635.32] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Adenoviruses have been critical in the development of the molecular approaches to brain tumors. They have been engineered to function as vectors for delivering therapeutic genes in gene therapy strategies, and as direct cytotoxic agents in oncolytic viral therapies. This review outlines the uses of adenoviruses in brain tumor therapy by examining clinical trials of adenovirus-mediated p53 gene therapy and by reviewing the application of two conditionally replicative adenoviruses (CRAds) ONYX-015 and Delta 24 in brain tumors. The potential clinical use of CRAds that deliver trangenes, particularly p53, is also discussed.
Collapse
Affiliation(s)
- Giacomo G Vecil
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | |
Collapse
|
37
|
Abstract
The potential use of adenoviruses in therapy against cancer has evoked a rapidly moving field of research. Unlike conventional gene therapy vectors, oncolytic adenoviruses retain the ability to replicate. However, replication is restricted as much as possible to tumor cells, with the aim of eliminating these cells through viral cytotoxicity. The two key issues are to improve the efficiency of virus replication and cell killing while ensuring the specificity of these activities for tumor cells. Wild-type adenoviruses as such may already be usable for cancer therapy. Strategies to further improve efficiency and specificity include the partial or complete removal of viral genes. The idea is that functions carried out by the corresponding gene products are not required for replication in tumor cells, but are needed in normal cells. Accordingly, the removal of genes encoding E1B-55 kDa or E1B-19 kDa, or the mutation of E1A may improve the selective killing of tumor cells. On the other hand, the overexpression of the adenovirus death protein (ADP) may enhance viral spread and oncolytic efficiency. Other strategies to improve the specific oncolytic activity of replicating adenoviruses have been pursued. For instance, some promoters are active specifically in tumor cells, and these promoters were introduced into the viral genome, to regulate essential viral genes. Moreover, replicating viruses were engineered to express toxic proteins or drug converters. A number of these viruses have been tested successfully using tumor xenografts in nude mice as a model system. An oncolytic adenovirus lacking the E1B-55 kDa gene product, termed dl1520 or ONYX015, was injected into squamous cell carcinomas of head and neck in phase II clinical trials, and the results were encouraging when chemotherapy was applied in parallel. In the future, further progress might be achieved on the level of virus constructs, but also by refining and adjusting simultaneous conventional therapies, and by standardizing the assessment of the clinical outcome. Recent progress has been made towards the use of replicating virus constructs in cancer therapy. The goal of these developments is to remove cancerous cells from patients with the help of viruses that selectively replicate in these cells. These viruses are generally termed oncolytic viruses. Some convenient properties of adenovirus make this virus particularly useful for this purpose. It infects a large number of human cell types, especially epithelial cells, which give rise to the vast majority of human malignancies. It can be grown easily and to high titers, and the creation of virus recombinants is well established. Finally, a large body of basic research has already been carried out on this virus, facilitating its manipulation. Various approaches to use adenovirus as a cancer drug have been reviewed (Alemany et al. 1999a, 2000; Curiel 2000; Galanis et al. 2001b; Gromeier 2001; Heise and Kirn 2000; Kirn 2000a; Kirn et al. 2001; Kirn and McCormick 1996; Smith and Chiocca 2000; Sunamura 2000; Wells 2000; Wodarz 2001). The aim of this chapter is to provide an integrated overview of these strategies.
Collapse
Affiliation(s)
- M Dobbelstein
- Institut für Virologie, Philipps-Universität Marburg, Robert Koch Str. 17, 35037 Marburg, Germany.
| |
Collapse
|
38
|
Abstract
Despite advances in current treatment modalities, the clinical outcome of gastric cancer remains dismal. New treatment modalities are urgently required to improve the prognosis of patients with gastric cancer. Cancer gene therapy and virotherapy comprise a potential category of new therapeutics and will be discussed in this review. To date, various gene therapy strategies have been developed, but first clinical trials reported only limited therapeutic efficacy as a result of limited gene transfer efficiency. Consequently, targeted viral vectors for enhanced delivery of transgenes to tumor cells and replicative viral systems designed to replicate selectively in malignant tissue were developed. Replication-selective oncolytic viral vectors have the advantage over non-replicative systems to cause pronounced bystander effect via self-perpetuating infection of adjacent cells after cytolysis of primary targeted cells. So far, clinical studies on virotherapy showed encouraging results; especially promising are combinations of virotherapy with current modes of treatment like chemo- and radiotherapy, or insertion of therapeutic genes in the viral genome such as combination with enzyme-prodrug therapy. Further research aiming to enhance anti-tumor efficacy and to improve selectivity of infection and replication, will eventually lead to full realization of the therapeutic potential of (replicating) viral vector systems for gastric cancer.
Collapse
|
39
|
Rein DT, Breidenbach M, Nettelbeck DM, Kawakami Y, Siegal GP, Huh WK, Wang M, Hemminki A, Bauerschmitz GJ, Yamamoto M, Adachi Y, Takayama K, Dall P, Curiel DT. Evaluation of tissue-specific promoters in carcinomas of the cervix uteri. J Gene Med 2004; 6:1281-9. [PMID: 15368588 DOI: 10.1002/jgm.606] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Gene therapy is a novel approach for treatment of patients with advanced, recurrent, or metastatic cervical cancer. One effective way to direct transgene expression to specific tissues or tumors is the use of tissue-specific-promoters (TSPs). In the context of adenovirus (Ad)-mediated cancer gene therapy it is rational to choose a TSP which is highly expressed in the tumor but has potentially low activity in non-tumor cells, especially the liver. In this study, we have investigated several promoters which fulfill these criteria. Candidate cervical cancer specific TSPs include promoters of the genes for secretory leukoprotease inhibitor (SLPI), cyclooxygenase-2 (COX-2), Midkine (MK), vascular endothelial growth factor receptor type 1 (flt-1), vascular endothelial growth factor (VEGF), Survivin and the receptor for chemokine SDS-1 (CXCR4). METHODS To evaluate the specific gene expression of the different promoters in the context of cervical cancer, we constructed a panel of E1-deleted Ads that express luciferase under the control of the promoters of interest. We investigated various established cervical cancer cell lines, as well as purified primary cancer cells and normal control cells from the cervix uteri. RESULTS In all cell lines tested, promoters for MK, VEGF and CXCR4 showed the highest activity. Both MK and VEGF promoters also resulted in a high activity in primary cervical cancer cells. Interestingly, gene expression profiles correlate with luciferase activity in both cell lines and primary cancer samples. CONCLUSIONS Our study demonstrates that the promoters for MK and VEGF are active in cervical cancer. We believe that both promoters can be successfully employed as TSPs for gene therapy targeted to cervical cancer.
Collapse
Affiliation(s)
- Daniel T Rein
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Drosten M, Pützer BM. Gene therapeutic approaches for medullary thyroid carcinoma treatment. J Mol Med (Berl) 2003; 81:411-9. [PMID: 12811413 DOI: 10.1007/s00109-003-0455-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2003] [Accepted: 05/21/2003] [Indexed: 11/25/2022]
Abstract
Medullary thyroid carcinoma (MTC), a neoplasm of thyroid C-cells, is characterized by dominant activating mutations in the RET proto-oncogene. Currently therapy is restricted to surgical removal of all neoplastic tissue lacking alternative forms of treatment such as chemotherapy or radiotherapy. Therefore MTC is a particularly attractive target for gene therapeutic approaches. Many promising gene therapy strategies have been used in various animal models of MTC, showing enhanced antitumoral efficacy, and these will hopefully extend our current standard of care in the future. These approaches can tentatively be subdivided into four groups: (a) Inhibition of oncogenic RET signaling, (b) suicide gene therapy, (c) immunotherapy, and (d) combination of immunotherapy and suicide approaches. To block oncogenic signal transduction dominant-negative RET mutants were delivered into tumor cells and found to possess strong antineoplastic activity, including tumor growth suppression and increased animal survival. Suicide gene therapeutic approaches applied to MTC treatment featured either gene transfer of herpes simplex virus thymidine kinase with concomitant application of ganciclovir or delivery of nitric oxide synthase II. Here antitumor effects were attributed to the occurrence of substantial bystander activities. Immunotherapy approaches comprised stimulation of immune response by delivery of interleukin 2 or 12. Finally, treatment with herpes simplex virus thymidine kinase/ganciclovir in combination with interleukin 2 was found to be superior over either treatment alone. This review discusses the various gene therapeutic approaches applied to MTC treatment in detail, gives an overview on the diverse vector systems used to achieve efficient transduction of thyroid cancer cells, and points out the strategies employed to accomplish target cell selective gene expression thereby contributing to enhanced safety of gene therapy for MTC
Collapse
Affiliation(s)
- Matthias Drosten
- Center for Cancer Research and Cancer Therapy, Institute of Molecular Biology, University of Essen Medical School, Hufelandstrasse 55, 45122 Essen, Germany
| | | |
Collapse
|
41
|
Noya F, Balagué C, Banerjee NS, Curiel DT, Broker TR, Chow LT. Activation of adenovirus early promoters and lytic phase in differentiated strata of organotypic cultures of human keratinocytes. J Virol 2003; 77:6533-40. [PMID: 12743310 PMCID: PMC155017 DOI: 10.1128/jvi.77.11.6533-6540.2003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human oncolytic adenoviruses have been used in clinical trials targeting cancers of epithelial origin. To gain a better understanding of the infectious cycle of adenovirus in normal human squamous tissues, we examined the viral infection process in organotypic cultures of primary human keratinocytes. We show that for the infection to occur, wounding of the epithelium is required. In addition, infection appears to initiate at the basal or parabasal cells that express the high-affinity coxsackievirus-adenovirus receptor, CAR, whereas the productive phase takes place in differentiated cells. This is due, at least in part, to the differentiation-dependent activation of the E1A and E2A early promoters and E4 promoters. We also show that adenovirus infection triggers a response mediated by the abnormal accumulation of cyclin E and p21cip1 proteins similar to the one previously observed in human papillomavirus-infected tissues. However, the virus seems to be able to overcome it, at least partially.
Collapse
Affiliation(s)
- Francisco Noya
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 35294-0005, USA
| | | | | | | | | | | |
Collapse
|
42
|
Lam JT, Bauerschmitz GJ, Kanerva A, Barker SD, Straughn JM, Wang M, Barnes MN, Blackwell JL, Siegal GP, Alvarez RD, Curiel DT, Hemminki A. Replication of an integrin targeted conditionally replicating adenovirus on primary ovarian cancer spheroids. Cancer Gene Ther 2003; 10:377-87. [PMID: 12719707 DOI: 10.1038/sj.cgt.7700578] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Replication competent viruses hold promise for treatment of advanced cancers resistant to available therapeutic modalities. Although preliminary clinical results have substantiated their efficacy, preclinical development of these novel approaches is limited by assay substrates. The evaluation of candidate agents could be confounded by differences between primary tumor cells and tumor cell lines, as discordance in the levels of surface receptors relevant for viral entry has been reported. Since primary tumor cells are difficult to analyze ex vivo for longitudinal observation of virus replication, we developed three-dimensional aggregates or spheroids of unpassaged and purified ovarian cancer cells as a means for prolonging primary tumor cell viability and as a three-dimensional in vitro model for replicative viral infection. Ovarian cancer cells purified from ascites samples were sustained for 30 days while retaining the infection profile with tropism modified and unmodified adenoviruses (Ads). Cell line and primary cell spheroids were used to quantitate the replication and oncolytic potency of replicative Ads in preclinical testing for human ovarian cancer trials. Therefore, spheroids provide a method to sustain purified unpassaged primary ovarian cancer cells for extended periods and to allow evaluation of replicative viruses in a three-dimensional model.
Collapse
Affiliation(s)
- John T Lam
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rainov NG, Ren H. Oncolytic viruses for treatment of malignant brain tumours. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 88:113-23. [PMID: 14531569 DOI: 10.1007/978-3-7091-6090-9_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Wild type viruses have been known for decades for their capability to destroy malignant tumour cells upon infection and intracellular replication. Genetic engineering of such viruses was, however, only recently done in an attempt to improve their utility as biological anticancer agents. Wild type or recombinant viruses able to selectively destroy tumour cells while sparing normal tissue are known as oncolytic viruses. Most oncolytic viruses currently investigated in clinical trials are derived from adenovirus (AV) or herpes simplex virus type I (HSVI). More than 300 patients with solid tumours were now treated in clinical trials with oncolytic viruses, and in most cases virus was administered directly into the tumour mass. About 10% of the above patients had recurrent malignant glioma. Total intratumoral doses of up to 2 x 10(12) virus particles were well tolerated, and in general no severe side effects resulted from the clinical use of oncolytic AV and HSVI, either in the brain or in the rest of the body. Encouraging anti-tumoral activity was demonstrated in some types of tumours treated locally with oncolytic viruses, and systemic chemotherapy was found to potentiate the anti-tumour effect of virus mediated oncolysis. In malignant glioma, standard gene therapy approaches employing non-replicating virus vectors failed to demonstrate significant benefit in clinical studies. Therapy with oncolytic viruses seems to hold more promise in early clinical trials than gene therapy with non-replicating virus vectors. However, further major advancements in virus designs, application modalities, and understanding of the interactions of the host's immune system with the virus are clearly needed before oncolytic virus therapy of malignant brain tumours can be introduced to clinical practice.
Collapse
Affiliation(s)
- N G Rainov
- Department of Neurological Science, University of Liverpool, UK.
| | | |
Collapse
|
44
|
Abstract
Although the cytotoxic effects of viruses are usually viewed in terms of pathogenicity, it is possible to harness this activity for therapeutic purposes. Viral genomes are highly versatile, and can be modified to direct their cytotoxicity towards cancer cells. These viruses are known as oncolytic viruses. How are viruses engineered to become tumour specific, and can they be used to safely treat cancer in humans?
Collapse
Affiliation(s)
- E Antonio Chiocca
- Molecular Neuro-Oncology Laboratories, Neurosurgery Service, Massachusetts General Hospital, East Building, 13th Street, Bldg 149, Charlestown, Massachusetts 02129, USA. chioccahelix.mgh.harvard.edu
| |
Collapse
|
45
|
Kruyt FAE, Curiel DT. Toward a new generation of conditionally replicating adenoviruses: pairing tumor selectivity with maximal oncolysis. Hum Gene Ther 2002; 13:485-95. [PMID: 11874627 DOI: 10.1089/10430340252809784] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Conditionally replicating adenoviruses (CRADs) represent a promising new platform for the treatment of cancer. CRADs have been demonstrated to kill tumor cells when other therapies fail, indicating that their antitumor properties are complementary to, and distinct from, those of standard treatments such as chemotherapy and radiation. In clinic trials CRADs have shown encouraging results, demonstrating mild side effects when administered at high doses and via different routes, including intratumorally, intraperitoneally, and intravenously. Tumor-selective replication has been detected, although as a single agent the efficacy appears to be limited. Interestingly, combined treatment with radiation or chemotherapy has been found to enhance CRAD efficacy considerably. To date, the molecular mechanisms underlying adenovirus-mediated oncolysis, and the way in which chemotherapy enhances oncolysis, are not well understood. A fuller knowledge of these processes will open up new strategies to improve the cell-killing potential of CRADs. Here, we discuss several possibilities that may lead to CRADs with enhanced oncolytic activity. These approaches include strategies to functionally couple tumor targeting and optimal oncolytic activity, and ways to further increase tumor cell disruption at later stages of infection to facilitate the spreading of virus throughout the tumor mass. In addition, improved methods to evaluate the efficacy of these agents in animal models, and in the clinic, will be required to systematically test and optimize CRAD efficacy, also taking into account the influence of tumor characteristics and the administration route.
Collapse
Affiliation(s)
- Frank A E Kruyt
- Division of Gene Therapy, Department of Medical Oncology, VU University Medical Center, NL-1007 MB Amsterdam, The Netherlands
| | | |
Collapse
|