1
|
Jalšić L, Lytvyn V, Elahi SM, Hrapovic S, Nassoury N, Chahal PS, Gaillet B, Gilbert R. Inducible HEK293 AAV packaging cell lines expressing Rep proteins. Mol Ther Methods Clin Dev 2023; 30:259-275. [PMID: 37560197 PMCID: PMC10407821 DOI: 10.1016/j.omtm.2023.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 07/11/2023] [Indexed: 08/11/2023]
Abstract
Packaging or producer cell lines for scalable recombinant adeno-associated virus (rAAV) production have been notoriously difficult to create due in part to the cytostatic nature of the Rep proteins required for AAV production. The most difficult challenge being creating AAV packaging cell lines using HEK293 parental cells, currently the best mammalian platform for rAAV production due to the constitutive expression of E1A in HEK293 cells, a key REP transcription activator. Using suspension and serum-free media adapted HEK293SF carrying a gene expression regulation system induced by addition of cumate and coumermycin, we were able to create REP-expressing AAV packaging cells. This was achieved by carefully choosing two of the AAV Rep proteins (Rep 40 and 68), using two inducible promoters with different expression levels and integrating into the cells through lentiviral vector transduction. Three of our best clones produced rAAV titers comparable to titers obtained by standard triple plasmid transfection of their parental cells. These clones were stable for up to 7 weeks under continuous cultures condition. rAAV production from one clone was also validated at scale of 1 L in a wave bioreactor using serum-free suspension culture.
Collapse
Affiliation(s)
- Lovro Jalšić
- Département de Génie Chimique, Université Laval, Québec, QC G1V0A6, Canada
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC H4P 2R2, Canada
| | - Viktoria Lytvyn
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC H4P 2R2, Canada
| | - Seyyed Mehdy Elahi
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC H4P 2R2, Canada
| | - Sabahudin Hrapovic
- Advanced Biomaterials and Chemical Synthesis Team, Aquatic and Crop Resource Development Research Centre, National Research Council Canada, Montréal, QC H4P 2R2, Canada
| | - Nasha Nassoury
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC H4P 2R2, Canada
| | - Parminder Singh Chahal
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC H4P 2R2, Canada
| | - Bruno Gaillet
- Département de Génie Chimique, Université Laval, Québec, QC G1V0A6, Canada
| | - Rénald Gilbert
- Département de Génie Chimique, Université Laval, Québec, QC G1V0A6, Canada
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC H4P 2R2, Canada
- Department of Bioengineering, McGill University, Montréal, QC H3A 0E9 Canada
| |
Collapse
|
2
|
Clinically Explored Virus-Based Therapies for the Treatment of Recurrent High-Grade Glioma in Adults. Biomedicines 2021; 9:biomedicines9020138. [PMID: 33535555 PMCID: PMC7912718 DOI: 10.3390/biomedicines9020138] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
As new treatment modalities are being explored in neuro-oncology, viruses are emerging as a promising class of therapeutics. Virotherapy consists of the introduction of either wild-type or engineered viruses to the site of disease, where they exert an antitumor effect. These viruses can either be non-lytic, in which case they are used to deliver gene therapy, or lytic, which induces tumor cell lysis and subsequent host immunologic response. Replication-competent viruses can then go on to further infect and lyse neighboring glioma cells. This treatment paradigm is being explored extensively in both preclinical and clinical studies for a variety of indications. Virus-based therapies are advantageous due to the natural susceptibility of glioma cells to viral infection, which improves therapeutic selectivity. Furthermore, lytic viruses expose glioma antigens to the host immune system and subsequently stimulate an immune response that specifically targets tumor cells. This review surveys the current landscape of oncolytic virotherapy clinical trials in high-grade glioma, summarizes preclinical experiences, identifies challenges associated with this modality across multiple trials, and highlights the potential to integrate this therapeutic strategy into promising combinatory approaches.
Collapse
|
3
|
Hajda J, Lehmann M, Krebs O, Kieser M, Geletneky K, Jäger D, Dahm M, Huber B, Schöning T, Sedlaczek O, Stenzinger A, Halama N, Daniel V, Leuchs B, Angelova A, Rommelaere J, Engeland CE, Springfeld C, Ungerechts G. A non-controlled, single arm, open label, phase II study of intravenous and intratumoral administration of ParvOryx in patients with metastatic, inoperable pancreatic cancer: ParvOryx02 protocol. BMC Cancer 2017; 17:576. [PMID: 28851316 PMCID: PMC5574242 DOI: 10.1186/s12885-017-3604-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/24/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metastatic pancreatic cancer has a dismal prognosis, with a mean six-month progression-free survival of approximately 50% and a median survival of about 11 months. Despite intensive research, only slight improvements of clinical outcome could be achieved over the last decades. Hence, new and innovative therapeutic strategies are urgently required. ParvOryx is a drug product containing native parvovirus H-1 (H-1PV). Since H-1PV was shown to exert pronounced anti-neoplastic effects in pre-clinical models of pancreatic cancer, the drug appears to be a promising candidate for treatment of this malignancy. METHODS ParvOryx02 is a non-controlled, single arm, open label, dose-escalating, single center trial. In total seven patients with pancreatic cancer showing at least one hepatic metastasis are to be treated with escalating doses of ParvOryx according to the following schedule: i) 40% of the total dose infused intravenously in equal fractions on four consecutive days, ii) 60% of the total dose injected on a single occasion directly into the hepatic metastasis at varying intervals after intravenous infusions. The main eligibility criteria are: age ≥ 18 years, disease progression despite first-line chemotherapy, and at least one hepatic metastasis. Since it is the second trial within the drug development program, the study primarily explores safety and tolerability after further dose escalation of ParvOryx. The secondary objectives are related to the evaluation of certain aspects of anti-tumor activity and clinical efficacy of the drug. DISCUSSION This trial strongly contributes to the clinical development program of ParvOryx. The individual hazards for patients included in the current study and the environmental risks are addressed and counteracted adequately. Besides information on safety and tolerability of the treatment after further dose escalation, thorough evaluations of pharmacokinetics and intratumoral spread as well as proof-of-concept (PoC) in pancreatic cancer will be gained in the course of the trial. TRIAL REGISTRATION ClinicalTrials.gov-ID: NCT02653313 , Registration date: Dec. 4th, 2015.
Collapse
Affiliation(s)
- Jacek Hajda
- Coordination Centre for Clinical Trials, University Hospital Heidelberg, Marsilius-Arkaden, Tower West, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany.
| | - Monika Lehmann
- Coordination Centre for Clinical Trials, University Hospital Heidelberg, Marsilius-Arkaden, Tower West, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Ottheinz Krebs
- Oryx GmbH & Co KG, Marktplatz 1, 85598, Baldham, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry and Informatics, University Hospital Heidelberg, Marsilius-Arkaden, Tower West, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Karsten Geletneky
- Department of Neurosurgery, Klinikum Darmstadt, Grafenstraße 9, 64283, Darmstadt, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Michael Dahm
- Oryx GmbH & Co KG, Marktplatz 1, 85598, Baldham, Germany
| | - Bernard Huber
- Oryx GmbH & Co KG, Marktplatz 1, 85598, Baldham, Germany
| | - Tilman Schöning
- Central Pharmacy, University Hospital Heidelberg, Im Neuenheimer Feld 670, 69120, Heidelberg, Germany
| | - Oliver Sedlaczek
- Department of Radiology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Albrecht Stenzinger
- Department of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Niels Halama
- Tissue Imaging & Analysis Center (TIGA), University Heidelberg - BioQuant, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| | - Volker Daniel
- Institute of Immunology, Transplantation Immunology, University Hospital Heidelberg, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Barbara Leuchs
- Department of Applied Tumor Virology, German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Assia Angelova
- Department of Applied Tumor Virology, German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Jean Rommelaere
- Department of Applied Tumor Virology, German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Christine E Engeland
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Guy Ungerechts
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| |
Collapse
|
4
|
Foreman PM, Friedman GK, Cassady KA, Markert JM. Oncolytic Virotherapy for the Treatment of Malignant Glioma. Neurotherapeutics 2017; 14:333-344. [PMID: 28265902 PMCID: PMC5398989 DOI: 10.1007/s13311-017-0516-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Malignant glioma is the most common primary brain tumor and carries a grim prognosis, with a median survival of just over 14 months. Given the poor outcomes with standard-of-care treatments, novel treatment strategies are needed. The concept of virotherapy for the treatment of malignant tumors dates back more than a century and can be divided into replication-competent oncolytic viruses and replication-deficient viral vectors. Oncolytic viruses are designed to selectively target, infect, and replicate in tumor cells, while sparing surrounding normal brain. A host of oncolytic viruses has been evaluated in early phase human trials with promising safety results, but none has progressed to phase III trials. Despite the 25 years that has passed since the initial publication of genetically engineered oncolytic viruses for the treatment of glioma, much remains to be learned about the use of this therapy, including its mechanism of action, optimal treatment paradigm, appropriate targets, and integration with adjuvant agents. Oncolytic viral therapy for glioma remains promising and will undoubtedly impact the future of patient care.
Collapse
Affiliation(s)
- Paul M Foreman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory K Friedman
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Tao L, Reese TA. Making Mouse Models That Reflect Human Immune Responses. Trends Immunol 2017; 38:181-193. [PMID: 28161189 DOI: 10.1016/j.it.2016.12.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/19/2016] [Accepted: 12/30/2016] [Indexed: 02/08/2023]
Abstract
Humans are infected with a variety of acute and chronic pathogens over the course of their lives, and pathogen-driven selection has shaped the immune system of humans. The same is likely true for mice. However, laboratory mice we use for most biomedical studies are bred in ultra-hygienic environments, and are kept free of specific pathogens. We review recent studies that indicate that pathogen infections are important for the basal level of activation and the function of the immune system. Consideration of these environmental exposures of both humans and mice can potentially improve mouse models of human disease.
Collapse
Affiliation(s)
- Lili Tao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tiffany A Reese
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
6
|
Abstract
Report of the Working Group on Hygiene of the Gesellschaft für Versuchstierkunde–Society for Laboratory Animal Science (GV-SOLAS) GV-SOLAS Working Group on Hygiene: Werner Nicklas (Chairman), Felix R. Homberger, Brunhilde Illgen-Wilcke, Karin Jacobi, Volker Kraft, Ivo Kunstyr, Michael Mähler, Herbert Meyer & Gabi Pohlmeyer-Esch
Collapse
|
7
|
Akladios C, Aprahamian M. Virotherapy of digestive tumors with rodent parvovirus: overview and perspectives. Expert Opin Biol Ther 2016; 16:645-53. [PMID: 26855087 DOI: 10.1517/14712598.2016.1151492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Toolan's H-1 parvovirus (H-1PV) exerts a cytotoxic/oncolytic effect, predominantly mediated by its non-structural protein (NS1). This rat parvovirus is harmless, unlike other parvoviruses, and its antitumor potential may be useful to clinicians as its oncolytic action appears to be true in numerous non-digestive and digestive cancers. AREAS COVERED After a brief review of parvovirus genus and biology, we summarize the proposed mechanisms to explain the cytotoxicity of H-1PV to tumors which results in dysregulation of cell transcription, cell-cycle arrest, termination of cell replication, activation of cellular stress response and induction of cell death. Viral oncolysis induces a strong tumor-specific immune response leading to the recognition and elimination of minimal residual disease. As the action of H-1PV is not limited to the digestive tract, we initially analyse studies performed in non-digestive cancers such as glioma (as the virus is able to cross the blood brain barrier), and then focused more particularly on the results in digestive cancers. EXPERT OPINION Based on the results of studies showing little H-1PV toxicity to living bodies, we advocate for the use of the parvovirus in cancers such as melanoma, glioma and pancreatic ductal adenocarcinoma in addition to conventional chemotherapy.
Collapse
Affiliation(s)
- Cherif Akladios
- a Institut de Recherche contre les Cancers Digestifs , 1 place de l'hôpital, 67000 Strasbourg , France
| | - Marc Aprahamian
- a Institut de Recherche contre les Cancers Digestifs , 1 place de l'hôpital, 67000 Strasbourg , France
| |
Collapse
|
8
|
Angelova AL, Geletneky K, Nüesch JPF, Rommelaere J. Tumor Selectivity of Oncolytic Parvoviruses: From in vitro and Animal Models to Cancer Patients. Front Bioeng Biotechnol 2015; 3:55. [PMID: 25954743 PMCID: PMC4406089 DOI: 10.3389/fbioe.2015.00055] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/05/2015] [Indexed: 11/23/2022] Open
Abstract
Oncolytic virotherapy of cancer is among the innovative modalities being under development and especially promising for targeting tumors, which are resistant to conventional treatments. Presently, at least a dozen of viruses, belonging to nine different virus families, are being tested within the frames of various clinical studies in cancer patients. Continuously growing preclinical evidence showing that the autonomous rat parvovirus H-1 (H-1PV) is able to kill tumor cells that resist conventional treatments and to achieve a complete cure of various human tumors in animal models argues for its inclusion in the arsenal of oncolytic viruses with an especially promising bench to bedside translation potential. Oncolytic parvovirus safe administration to humans relies on the intrinsic preference of these agents for quickly proliferating, metabolically, and biochemically disturbed tumor versus normal cells (tumor selectivity or oncotropism). The present review summarizes and discusses (i) preclinical evidence of H-1PV innocuousness for normal cells and healthy tissues in vitro and in animals, respectively, (ii) toxicological assessments of H-1PV mono- or combined therapy in tumor-bearing virus-permissive animal models, as well as (iii) historical results of experimental infection of human cancer patients with H-1PV. Altogether, these data argue against a risk of H-1PV inducing significant toxic effects in human patients. This highly favorable safety profile allowed the translation of H-1PV preclinical research into a Phase I/IIa clinical trial being currently in progress.
Collapse
Affiliation(s)
- Assia L Angelova
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Karsten Geletneky
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany ; Department of Neurosurgery, University of Heidelberg , Heidelberg , Germany
| | - Jürg P F Nüesch
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Jean Rommelaere
- Infection and Cancer Program, Division of Tumor Virology, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
9
|
Novel permissive murine immunocompetent orthotopic colon carcinoma model for comparison of the antitumoral and safety profiles of three Adv-TKs. Gene Ther 2014; 21:975-83. [DOI: 10.1038/gt.2014.79] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 06/21/2014] [Accepted: 07/02/2014] [Indexed: 01/02/2023]
|
10
|
Huang LY, Halder S, Agbandje-McKenna M. Parvovirus glycan interactions. Curr Opin Virol 2014; 7:108-18. [PMID: 25047752 DOI: 10.1016/j.coviro.2014.05.007] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/27/2014] [Indexed: 12/30/2022]
Abstract
Members of the Parvoviridae utilize glycan receptors for cellular attachment and subsequent interactions determine transduction efficiency or pathogenic outcome. This review focuses on the identity of the glycan receptors utilized, their capsid binding footprints, and a discussion of the overlap of these sites with tropism, transduction, and pathogenicity determinants. Despite high sequence diversity between the different genera, most parvoviruses bind to negatively charged glycans, such as sialic acid and heparan sulfate, abundant on cell surface membranes. The capsid structure of these viruses exhibit high structural homology enabling common regions to be utilized for glycan binding. At the same time the sequence diversity at the common footprints allows for binding of different glycans or differential binding of the same glycan.
Collapse
Affiliation(s)
- Lin-Ya Huang
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Sujata Halder
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
11
|
Nüesch JPF, Rommelaere J. Tumor suppressing properties of rodent parvovirus NS1 proteins and their derivatives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:99-124. [PMID: 25001533 DOI: 10.1007/978-1-4471-6458-6_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer chemotherapy with monospecific agents is often hampered by the rapid development of tumor resistance to the drug used. Therefore, combination treatments aiming at several different targets are sought. Viral regulatory proteins, modified or not, appear ideal for this purpose because of their multimodal killing action against neoplastically transformed cells. The large nonstructural protein NS1 of rodent parvoviruses is an excellent candidate for an anticancer agent, shown to interfere specifically with cancer cell growth and survival. The present review describes the structure, functions, and regulation of the multifunctional protein NS1, its specific interference with cell processes and cell protein activities, and what is known so far about the mechanisms underlying NS1 interference with cancer growth. It further outlines prospects for the development of new, multimodal cancer toxins and their potential applications.
Collapse
Affiliation(s)
- Jürg P F Nüesch
- Program "Infection and Cancer", Division Tumor Virology (F010), Deutsches Krebsforschungszentrum/German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, D-69120, Heidelberg, Germany,
| | | |
Collapse
|
12
|
Lavie M, Struyf S, Stroh-Dege A, Rommelaere J, Van Damme J, Dinsart C. Capacity of wild-type and chemokine-armed parvovirus H-1PV for inhibiting neo-angiogenesis. Virology 2013; 447:221-32. [PMID: 24210118 DOI: 10.1016/j.virol.2013.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 07/30/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
Abstract
Anti-angiogenic therapy has been recognized as a powerful potential strategy for impeding the growth of various tumors. However no major therapeutic effects have been observed to date, mainly because of the emergence of several resistance mechanisms. Among novel strategies to target tumor vasculature, some oncolytic viruses open up new prospects. In this context, we addressed the question whether the rodent parvovirus H-1PV can target endothelial cells. We show that cultures of human normal (HUVEC) and immortalized (KS-IMM) endothelial cells sustain an abortive viral cycle upon infection with H-1PV and are sensitive to H-1PV cytotoxicity. H-1PV significantly inhibits infected KS-IMM tumor growth. This effect may be traced back by the virus ability to both kill proliferating endothelial cells and inhibit VEGF production Recombinant H-1PV vectors can also transduce tumor cells with chemokines endowed with anti-angiogenesis properties, and warrant further validation for the treatment of highly vascularized tumors.
Collapse
Affiliation(s)
- Muriel Lavie
- Tumor Virology Division, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
13
|
NK-cell-dependent killing of colon carcinoma cells is mediated by natural cytotoxicity receptors (NCRs) and stimulated by parvovirus infection of target cells. BMC Cancer 2013; 13:367. [PMID: 23902851 PMCID: PMC3733944 DOI: 10.1186/1471-2407-13-367] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 07/29/2013] [Indexed: 12/30/2022] Open
Abstract
Background Investigating how the immune system functions during malignancies is crucial to developing novel therapeutic strategies. Natural killer (NK) cells, an important component of the innate immune system, play a vital role in immune defense against tumors and virus-infected cells. The poor survival rate in colon cancer makes it particularly important to develop novel therapeutic strategies. Oncolytic viruses, in addition to lysing tumor cells, may have the potential to augment antitumor immune responses. In the present study, we investigate the role of NK cells and how parvovirus H-1PV can modulate NK-cell mediated immune responses against colon carcinoma. Methods Human NK cells were isolated from the blood of healthy donors. The cytotoxicity and antibody-mediated inhibition of NK cells were measured in chromium release assays. Phenotypic assessment of colon cancer and dendritic cells was done by FACS. The statistical significance of the results was calculated with Student’s t test (*p <0.05; **, p < 0.01; ***, p < 0.001). Results We show that IL-2-activated human NK cells can effectively kill colon carcinoma cells. Killing of colon carcinoma cells by NK cells was further enhanced upon infection of the former cells with parvovirus H-1PV. H-1PV has potent oncolytic activity against various tumors, yet its direct killing effect on colon carcinoma cells is limited. The cytotoxicity of NK cells towards colon carcinoma cells, both mock- and H-1PV-infected, was found to be mostly mediated by a combination of natural cytotoxicity receptors (NCRs), namely NKp30, 44, and 46. Colon carcinoma cells displayed low to moderate expression of NK cell ligands, and this expression was modulated upon H-1PV infection. Lysates of H-1PV-infected colon carcinoma cells were found to increase MHC class II expression on dendritic cells. Conclusions Altogether, these data suggest that IL-2-activated NK cells actively kill colon carcinoma cells and that this killing is mediated by several natural cytotoxicity receptors (NCRs) in combination. Additionally, in association with parvovirus H-1PV, IL-2-activated NK cells have the potential to boost immune responses against colon cancer.
Collapse
|
14
|
Wennier ST, Liu J, McFadden G. Bugs and drugs: oncolytic virotherapy in combination with chemotherapy. Curr Pharm Biotechnol 2013; 13:1817-33. [PMID: 21740354 DOI: 10.2174/138920112800958850] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 09/18/2010] [Indexed: 12/16/2022]
Abstract
Single agent therapies are rarely successful in treating cancer, particularly at metastatic or end stages, and survival rates with monotherapies alone are generally poor. The combination of multiple therapies to treat cancer has already driven significant improvements in the standard of care treatments for many types of cancers. The first combination treatments exploited for cancer therapy involved the use of several cytotoxic chemotherapy agents. Later, with the development of more targeted agents, the use of novel, less toxic drugs, in combination with the more classic cytotoxic drugs has proven advantageous for certain cancer types. Recently, the combination of oncolytic virotherapy with chemotherapy has shown that the use of these two therapies with very distinct anti-tumor mechanisms may also lead to synergistic interactions that ultimately result in increased therapeutic effects not achievable by either therapy alone. The mechanisms of synergy between oncolytic viruses (OVs) and chemotherapeutic agents are just starting to be elucidated. It is evident, however, that the success of these OV-drug combinations depends greatly on the particular OV, the drug(s) selected, and the cancer type targeted. This review summarizes the different OV-drug combinations investigated to date, including the use of second generation armed OVs, which have been studied with the specific purpose of generating synergistic interactions with particular chemotherapy agents. The known mechanisms of synergy between these OV-drug combinations are also summarized. The importance of further investigating these mechanisms of synergy will be critical in order to maximize the therapeutic efficacy of OV-drug combination therapies in the future.
Collapse
Affiliation(s)
- Sonia Tusell Wennier
- Department of Molecular Genetics and Microbiology, University of Florida, 1600 SW Archer Rd, P.O. Box 100266 Gainesville, FL 32610, USA
| | | | | |
Collapse
|
15
|
Halder S, Nam HJ, Govindasamy L, Vogel M, Dinsart C, Salomé N, McKenna R, Agbandje-McKenna M. Production, purification, crystallization and structure determination of H-1 Parvovirus. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:1571-6. [PMID: 23192051 PMCID: PMC3509992 DOI: 10.1107/s1744309112045563] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 11/04/2012] [Indexed: 11/10/2022]
Abstract
Crystals of H-1 Parvovirus (H-1PV), an antitumor gene-delivery vector, were obtained for DNA-containing capsids and diffracted X-rays to 2.7 Å resolution using synchrotron radiation. The crystals belonged to the monoclinic space group P2(1), with unit-cell parameters a=255.4, b=350.4, c=271.6 Å, β=90.34°. The unit cell contained two capsids, with one capsid per crystallographic asymmetric unit. The H-1PV structure has been determined by molecular replacement and is currently being refined.
Collapse
Affiliation(s)
- Sujata Halder
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Hyun-Joo Nam
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Lakshmanan Govindasamy
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Michèle Vogel
- Research Program Infection and Cancer, Division of Tumor Virology, DKFZ F010 – Inserm U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Christiane Dinsart
- Research Program Infection and Cancer, Division of Tumor Virology, DKFZ F010 – Inserm U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Nathalie Salomé
- Research Program Infection and Cancer, Division of Tumor Virology, DKFZ F010 – Inserm U701, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
16
|
Sieben M, Schäfer P, Dinsart C, Galle PR, Moehler M. Activation of the human immune system via toll-like receptors by the oncolytic parvovirus H-1. Int J Cancer 2012; 132:2548-56. [PMID: 23151948 DOI: 10.1002/ijc.27938] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 10/05/2012] [Indexed: 01/09/2023]
Abstract
This study aimed to investigate the function of toll-like receptors (TLRs) during oncolytic parvovirus H-1 (H-1PV)-induced human immune responses. First, the role of TLRs in the activation of the NFκB transcription factor was characterized; second, the immunologic effects of H-1PV-induced tumor cell lysates (TCL) on human antitumor immune responses were evaluated. A human ex vivo model was used to study immune responses with dendritic cells (DCs). Human embryonic kidney cells (HEK293) transfected to stably express TLRs were used as potential human DC equivalents to further investigate the role of specific TLRs during immune activation. TLR3 and TLR9 were activated by H-1PV infection, which correlated with NFκB translocation to the nucleus and a reduced cytoplasmic IκB expression. Using a TLR-signaling reporter plasmid (pNiFty-Luc), NFκB activity was increased following H-1PV infection. In addition, human DCs coincubated with H-1PV-induced TCL demonstrated increased TLR3 and TLR9 expression. These data suggest that H-1PV-induced TCL stimulate human DCs at least in part through TLR-dependent signaling pathways. Thus, DC maturation occurred through exposure to H-1PV-induced TCL through TLR-signaling leading to NFκB-dependent activation of the adaptive immune system as indicated by the increased expression of CD86, TLR3 and TLR9. Furthermore, the transcription of various cytokines indicates the activation of immune response, therefore the production of the proinflammatory cytokine TNF-α was determined. Here, H-1PV-induced TCL significantly enhanced the TNF-α level by DCs after coculture. H-1PV oncolytic virotherapy enhances immune priming by different effects on DCs and generates antitumor immunity. These findings potentially offer a new approach to tumor therapy.
Collapse
Affiliation(s)
- Maike Sieben
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | | | | | | |
Collapse
|
17
|
Generation of an adenovirus-parvovirus chimera with enhanced oncolytic potential. J Virol 2012; 86:10418-31. [PMID: 22787235 DOI: 10.1128/jvi.00848-12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, our goal was to generate a chimeric adenovirus-parvovirus (Ad-PV) vector that combines the high-titer and efficient gene transfer of adenovirus with the anticancer potential of rodent parvovirus. To this end, the entire oncolytic PV genome was inserted into a replication-defective E1- and E3-deleted Ad5 vector genome. As we found that parvoviral NS expression inhibited Ad-PV chimera production, we engineered the parvoviral P4 early promoter, which governs NS expression, by inserting into its sequence tetracycline operator elements. As a result of these modifications, P4-driven expression was blocked in the packaging T-REx-293 cells, which constitutively express the tetracycline repressor, allowing high-yield chimera production. The chimera effectively delivered the PV genome into cancer cells, from which fully infectious replication-competent parvovirus particles were generated. Remarkably, the Ad-PV chimera exerted stronger cytotoxic activities against various cancer cell lines, compared with the PV and Ad parental viruses, while being still innocuous to a panel of tested healthy primary human cells. This Ad-PV chimera represents a novel versatile anticancer agent which can be subjected to further genetic manipulations in order to reinforce its enhanced oncolytic capacity through arming with transgenes or retargeting into tumor cells.
Collapse
|
18
|
Zeyaullah M, Patro M, Ahmad I, Ibraheem K, Sultan P, Nehal M, Ali A. Oncolytic viruses in the treatment of cancer: a review of current strategies. Pathol Oncol Res 2012; 18:771-81. [PMID: 22714538 DOI: 10.1007/s12253-012-9548-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 05/30/2012] [Indexed: 12/18/2022]
Abstract
Oncolytic viruses are live, replication-competent viruses that replicate selectively in tumor cells leading to the destruction of the tumor cells. Tumor-selective replicating viruses offer appealing advantages over conventional cancer therapy and are promising a new approach for the treatment of human cancer. The development of virotherapeutics is based on several strategies. Virotherapy is not a new concept, but recent technical advances in the genetic modification of oncolytic viruses have improved their tumor specificity, leading to the development of new weapons for the war against cancer. Clinical trials with oncolytic viruses demonstrate the safety and feasibility of an effective virotherapeutic approach. Strategies to overcome potential obstacles and challenges to virotherapy are currently being explored. Systemic administrations of oncolytic viruses will successfully extend novel treatment against a range of tumors. Combination therapy has shown some encouraging antitumor responses by eliciting strong immunity against established cancer.
Collapse
Affiliation(s)
- Md Zeyaullah
- Department of Microbiology, Faculty of Medicine, Omar Al-Mukhtar University, Al-Baida, Libya.
| | | | | | | | | | | | | |
Collapse
|
19
|
LuIII parvovirus selectively and efficiently targets, replicates in, and kills human glioma cells. J Virol 2012; 86:7280-91. [PMID: 22553327 DOI: 10.1128/jvi.00227-12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Because productive infection by parvoviruses requires cell division and is enhanced by oncogenic transformation, some parvoviruses may have potential utility in killing cancer cells. To identify the parvovirus(es) with the optimal oncolytic effect against human glioblastomas, we screened 12 parvoviruses at a high multiplicity of infection (MOI). MVMi, MVMc, MVM-G17, tumor virus X (TVX), canine parvovirus (CPV), porcine parvovirus (PPV), rat parvovirus 1A (RPV1A), and H-3 were relatively ineffective. The four viruses with the greatest oncolytic activity, LuIII, H-1, MVMp, and MVM-G52, were tested for the ability, at a low MOI, to progressively infect the culture over time, causing cell death at a rate higher than that of cell proliferation. LuIII alone was effective in all five human glioblastomas tested. H-1 progressively infected only two of five; MVMp and MVM-G52 were ineffective in all five. To investigate the underlying mechanism of LuIII's phenotype, we used recombinant parvoviruses with the LuIII capsid replacing the MVMp capsid or with molecular alteration of the P4 promoter. The LuIII capsid enhanced efficient replication and oncolysis in MO59J gliomas cells; other gliomas tested required the entire LuIII genome to exhibit enhanced infection. LuIII selectively infected glioma cells over normal glial cells in vitro. In mouse models, human glioblastoma xenografts were selectively infected by LuIII when administered intratumorally; LuIII reduced tumor growth by 75%. LuIII also had the capacity to selectively infect subcutaneous or intracranial gliomas after intravenous inoculation. Intravenous or intracranial LuIII caused no adverse effects. Intracranial LuIII caused no infection of mature mouse neurons or glia in vivo but showed a modest infection of developing neurons.
Collapse
|
20
|
Activation of a glioma-specific immune response by oncolytic parvovirus Minute Virus of Mice infection. Cancer Gene Ther 2012; 19:468-75. [PMID: 22539062 DOI: 10.1038/cgt.2012.20] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Rodent autonomous parvoviruses (PVs) are endowed with oncotropic properties and represent virotherapeutics with inherent oncolytic features. This work aimed to evaluate the capacity of Minute Virus of Mice (MVMp) to act as an adjuvant stimulating a mouse glioblastoma-specific immune response. MVMp was shown to induce cell death through apoptosis in glioma GL261 cells. Antigen-presenting cells (APCs) provide the initial cue for innate and adaptive immune responses, and thus MVMp-infected GL261 cells were tested for their ability to activate dendritic cells (DCs) and microglia (MG), two distinct cell types that are able to act as APCs. MG and discrete DC subsets were activated after co-culture with MVMp-infected glioma GL261 cells, as evidenced by upregulation of specific activation markers (CD80, CD86) and release of proinflammatory cytokines (tumor necrosis factor-α and interleukin-6). The in vivo analysis of immunodeficient and immunocompetent mice revealed a clear difference in their susceptibility to MVMp-mediated tumor suppression. Immunocompetent mice were fully protected from tumor outgrowth of GL261 cells infected ex vivo with MVMp. In contrast, immunodeficient animals were less competent for MVMp-dependent tumor inhibition, with only 20% of the recipients being protected, arguing for an additional immune component to allow full tumor suppression. In keeping with this conclusion, immunocompetent mice engrafted with MVMp-infected glioma cells developed a level of anti-tumor immunity with isolated splenocytes producing elevated levels of interferon-γ. In rechallenge experiments using uninfected GL261 cells, we could show complete protection against the tumor, arguing for the induction of a T-cell-mediated, tumor-specific, long-term memory response. These findings indicate that the anticancer effect of PVs can be traced back not only for their direct oncolytic effect, but also to their ability to break tumor tolerance.
Collapse
|
21
|
Geletneky K, Huesing J, Rommelaere J, Schlehofer JR, Leuchs B, Dahm M, Krebs O, von Knebel Doeberitz M, Huber B, Hajda J. Phase I/IIa study of intratumoral/intracerebral or intravenous/intracerebral administration of Parvovirus H-1 (ParvOryx) in patients with progressive primary or recurrent glioblastoma multiforme: ParvOryx01 protocol. BMC Cancer 2012; 12:99. [PMID: 22436661 PMCID: PMC3425127 DOI: 10.1186/1471-2407-12-99] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 03/21/2012] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The treatment of patients with malignant brain tumors remains a major oncological problem. The median survival of patients with glioblastoma multiforme (GBM), the most malignant type, is only 15 months after initial diagnosis and even less after tumor recurrence. Improvements of standard treatment including surgery and radio-chemotherapy have not lead to major improvements. Therefore, alternative therapeutics such as oncolytic viruses that specifically target and destroy cancer cells are under investigation. Preclinical data of oncolytic parvovirus H-1 (H-1PV) infection of glioma cells demonstrated strong cytotoxic and oncosuppressing effects, leading to a phase I/IIa trial of H-1PV in patients with recurrent GBM (ParvOryx01). ParvOryx01 is the first trial with a replication competent oncolytic virus in Germany. METHODS ParvOryx01 is an open, non-controlled, two groups, intra-group dose escalation, single center, phase I/IIa trial. 18 patients with recurrent GBM will be treated in 2 groups of 9 patients each. Treatment group 1 will first receive H-1PV by intratumoral injection and second by administration into the walls of the tumor cavity during tumor resection. In treatment group 2 the virus will initially be injected intravenously and afterwards, identical to group 1, into the surrounding brain tissue during tumor removal. Main eligibility criteria are: age of 18 years, unifocal recurrent GBM, amenable to complete or subtotal resection. Dose escalation will be based on the Continual Reassessment Method. The primary objective of the trial is local and systemic safety and tolerability and to determine the maximum tolerated dose (MTD). Secondary objectives are proof of concept (PoC) and Progression-free Survival (PFS) up to 6 months. DISCUSSION This is the first trial with H-1PV in patients with recurrent GBM. The risks for the participants appear well predictable and justified. Furthermore, ParvOryx01 will be the first assessment of combined intratumoral and intravenous application of an oncolytic virus. Due to its study design the trial will not only generate data on the local effect of H-1PV but it will also investigate the penetration of H-1PV into the tumor after systemic delivery and obtain safety data from systemic delivery possibly supporting clinical trials with H-1PV in other, non-CNS malignancies. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01301430.
Collapse
|
22
|
Moralès O, Richard A, Martin N, Mrizak D, Sénéchal M, Miroux C, Pancré V, Rommelaere J, Caillet-Fauquet P, de Launoit Y, Delhem N. Activation of a helper and not regulatory human CD4+ T cell response by oncolytic H-1 parvovirus. PLoS One 2012; 7:e32197. [PMID: 22359669 PMCID: PMC3281136 DOI: 10.1371/journal.pone.0032197] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 01/24/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND H-1 parvovirus (H-1 PV), a rodent autonomous oncolytic parvovirus, has emerged as a novel class of promising anticancer agents, because of its ability to selectively find and destroy malignant cells. However, to probe H-1 PV multimodal antitumor potential one of the major prerequisites is to decipher H-1 PV direct interplay with human immune system, and so prevent any risk of impairment. METHODOLOGY/PRINCIPAL FINDINGS Non activated peripheral blood mononuclear cells (PBMCs) are not sensitive to H-1 PV cytotoxic effect. However, the virus impairs both activated PBMC proliferation ability and viability. This effect is related to H-1 PV infection as evidenced by Western blotting detection of H-1 PV main protein NS1. However, TCID50 experiments did not allow newly generated virions to be detected. Moreover, flow cytometry has shown that H-1 PV preferentially targets B lymphocytes. Despite seeming harmful at first sight, H-1 PV seems to affect very few NK cells and CD8+ T lymphocytes and, above all, clearly does not affect human neutrophils and one of the major CD4+ T lymphocyte subpopulation. Very interestingly, flow cytometry analysis and ELISA assays proved that it even activates human CD4+ T cells by increasing activation marker expression (CD69 and CD30) and both effective Th1 and Th2 cytokine secretion (IL-2, IFN-γ and IL-4). In addition, H-1 PV action does not come with any sign of immunosuppressive side effect. Finally, we have shown the efficiency of H-1 PV on xenotransplanted human nasopharyngeal carcinoma, in a SCID mouse model reconstituted with human PBMC. CONCLUSIONS/SIGNIFICANCE Our results show for the first time that a wild-type oncolytic virus impairs some immune cell subpopulations while directly activating a Helper CD4+ T cell response. Thus, our data open numerous gripping perspectives of investigation and strongly argue for the use of H-1 PV as an anticancer treatment.
Collapse
Affiliation(s)
- Olivier Moralès
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Audrey Richard
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Nathalie Martin
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Dhafer Mrizak
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Magalie Sénéchal
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Céline Miroux
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Véronique Pancré
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Jean Rommelaere
- Tumor Virology, Research Program Infection and Cancer, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Perrine Caillet-Fauquet
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Yvan de Launoit
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
| | - Nadira Delhem
- Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
- * E-mail:
| |
Collapse
|
23
|
Abstract
Viral infections of laboratory mice have considerable impact on research results, and prevention of such infections is therefore of crucial importance. This chapter covers infections of mice with the following viruses: herpesviruses, mousepox virus, murine adenoviruses, polyomaviruses, parvoviruses, lactate dehydrogenase-elevating virus, lymphocytic choriomeningitis virus, mammalian orthoreovirus serotype 3, murine hepatitis virus, murine norovirus, murine pneumonia virus, murine rotavirus, Sendai virus, and Theiler’s murine encephalomyelitis virus. For each virus, there is a description of the agent, epizootiology, clinical symptoms, pathology, methods of diagnosis and control, and its impact on research.
Collapse
|
24
|
Janus LM, Bleich A. Coping with parvovirus infections in mice: health surveillance and control. Lab Anim 2012; 46:14-23. [DOI: 10.1258/la.2011.011025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Parvoviruses of mice, minute virus of mice (MVM) and mouse parvovirus (MPV), are challenging pathogens to eradicate from laboratory animal facilities. Due to the impediment on rodent-based research, recent studies have focused on the assessment of re-derivation techniques and parvoviral potential to induce persistent infections. Summarizing recent data, this review gives an overview on studies associated with parvoviral impact on research, diagnostic methods, parvoviral persistence and re-derivation techniques, demonstrating the complex nature of parvovirus infection in mice and unfolding the challenge of controlling parvovirus infections in laboratory animal facilities.
Collapse
Affiliation(s)
- Lydia M Janus
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
25
|
Adeno-associated virus type 2 modulates the host DNA damage response induced by herpes simplex virus 1 during coinfection. J Virol 2011; 86:143-55. [PMID: 22013059 DOI: 10.1128/jvi.05694-11] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Adeno-associated virus type 2 (AAV2) is a human parvovirus that relies on a helper virus for efficient replication. Herpes simplex virus 1 (HSV-1) supplies helper functions and changes the environment of the cell to promote AAV2 replication. In this study, we examined the accumulation of cellular replication and repair proteins at viral replication compartments (RCs) and the influence of replicating AAV2 on HSV-1-induced DNA damage responses (DDR). We observed that the ATM kinase was activated in cells coinfected with AAV2 and HSV-1. We also found that phosphorylated ATR kinase and its cofactor ATR-interacting protein were recruited into AAV2 RCs, but ATR signaling was not activated. DNA-PKcs, another main kinase in the DDR, was degraded during HSV-1 infection in an ICP0-dependent manner, and this degradation was markedly delayed during AAV2 coinfection. Furthermore, we detected phosphorylation of DNA-PKcs during AAV2 but not HSV-1 replication. The AAV2-mediated delay in DNA-PKcs degradation affected signaling through downstream substrates. Overall, our results demonstrate that coinfection with HSV-1 and AAV2 provokes a cellular DDR which is distinct from that induced by HSV-1 alone.
Collapse
|
26
|
Paglino J, Tattersall P. The parvoviral capsid controls an intracellular phase of infection essential for efficient killing of stepwise-transformed human fibroblasts. Virology 2011; 416:32-41. [PMID: 21600623 DOI: 10.1016/j.virol.2011.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 04/01/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
Abstract
Members of the rodent subgroup of the genus Parvovirus exhibit lytic replication and spread in many human tumor cells and are therefore attractive candidates for oncolytic virotherapy. However, the significant variation in tumor tropism observed for these viruses remains largely unexplained. We report here that LuIII kills BJ-ELR 'stepwise-transformed' human fibroblasts efficiently, while MVM does not. Using viral chimeras, we mapped this property to the LuIII capsid gene, VP2, which is necessary and sufficient to confer the killer phenotype on MVM. LuIII VP2 facilitates a post-entry, pre-DNA-amplification step early in the life cycle, suggesting the existence of an intracellular moiety whose efficient interaction with the incoming capsid shell is critical to infection. Thus targeting of human cancers of different tissue-type origins will require use of parvoviruses with capsids that effectively make this critical interaction.
Collapse
Affiliation(s)
- Justin Paglino
- Department of Laboratory Medicine, Yale University Medical School, New Haven, CT 06520, USA
| | | |
Collapse
|
27
|
Mincberg M, Gopas J, Tal J. Minute virus of mice (MVMp) infection and NS1 expression induce p53 independent apoptosis in transformed rat fibroblast cells. Virology 2011; 412:233-43. [DOI: 10.1016/j.virol.2010.12.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 09/10/2010] [Accepted: 12/20/2010] [Indexed: 10/18/2022]
|
28
|
Oncolytic virotherapy of gynecologic malignancies. Gynecol Oncol 2011; 120:302-10. [DOI: 10.1016/j.ygyno.2010.10.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/21/2010] [Accepted: 10/24/2010] [Indexed: 11/20/2022]
|
29
|
Bhat R, Dempe S, Dinsart C, Rommelaere J. Enhancement of NK cell antitumor responses using an oncolytic parvovirus. Int J Cancer 2011; 128:908-19. [PMID: 20473905 DOI: 10.1002/ijc.25415] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells play a vital role in the rejection of tumors. Pancreatic ductal adenocarcinoma (PDAC), however, remains a poor prognosis malignancy, due to its resistance to radio- and chemotherapy, and low immunogenicity. We demonstrate here that IL-2-activated human NK cells are able to kill PDAC cells. Currently, novel strategies are being pursued to combat PDAC. In this regard, oncolytic viruses, in addition to killing tumor cells, may also have the potential to augment antitumor immune responses. We found that, besides having an intrinsic oncolytic activity, parvovirus H-1PV is able to enhance NK cell-mediated killing of PDAC cells. Our results show that H-1PV infection of Panc-1 cells increases NK cell capacity to release IFN-γ, TNF-α and MIP-1α/β. Multiple activating receptors are involved in the NK cell-mediated killing of Panc-1 cells. Indeed, blocking of the natural cytotoxicity receptors-NKp30, 44 and 46 in combination, and NKG2D and DNAM1 alone inhibit the killing of Panc-1 cells. Interestingly, H-1PV infection of Panc-1 cells overcomes the part of inhibitory effects suggesting that parvovirus may induce additional NK cell ligands on Panc-1 cells. The enhanced sensitivity of H-1PV-infected PDAC cells to NK cell-dependent killing could be traced back to the upregulation of the DNAM-1 ligand, CD155 and to the downregulation of MHC class I expression. Our data suggests that NK cells display antitumor potential against PDAC and that H-1PV-based oncolytic immunotherapy could further boost NK cell-mediated immune responses and help to develop a combinatorial therapeutic approach against PDAC.
Collapse
Affiliation(s)
- Rauf Bhat
- Division of Tumor Virology, F010, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | |
Collapse
|
30
|
Bourke MG, Salwa S, Harrington KJ, Kucharczyk MJ, Forde PF, de Kruijf M, Soden D, Tangney M, Collins JK, O'Sullivan GC. The emerging role of viruses in the treatment of solid tumours. Cancer Treat Rev 2011; 37:618-32. [PMID: 21232872 DOI: 10.1016/j.ctrv.2010.12.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/04/2010] [Accepted: 12/07/2010] [Indexed: 12/13/2022]
Abstract
There is increasing optimism for the use of non-pathogenic viruses in the treatment of many cancers. Initial interest in oncolytic virotherapy was based on the observation of an occasional clinical resolution of a lymphoma after a systemic viral infection. In many cancers, by comparison with normal tissues, the competency of the cellular anti-viral mechanism is impaired, thus creating an exploitable difference between the tumour and normal cells, as an unimpeded viral proliferation in cancer cells is eventually cytocidal. In addition to their oncolytic capability, these particular viruses may be engineered to facilitate gene delivery to tumour cells to produce therapeutic effects such as cytokine secretion and anti -tumour immune responses prior to the eventual cytolysis. There is now promising clinical experience with these viral strategies, particularly as part of multimodal studies, and already several clinical trials are in progress. The limitations of standard cancer chemotherapies, including their lack of specificity with consequent collateral toxicity and the development of cross-resistance, do not appear to apply to viral-based therapies. Furthermore, virotherapy frequently restores chemoradiosensitivity to resistant tumours and has also demonstrated efficacy against cancers that historically have a dismal prognosis. While there is cause for optimism, through continued improvements in the efficiency and safety of systemic delivery, through the emergence of alternative viral agents and through favourable clinical experiences, clinical trials as part of multimodal protocols will be necessary to define clinical utility. Significant progress has been made and this is now a major research area with an increasing annual bibliography.
Collapse
Affiliation(s)
- M G Bourke
- Cork Cancer Research Centre, Leslie C. Quick Jnr. Laboratory, Biosciences Institute, University College Cork, Ireland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Novel adenovirus-based helper system to support production of recombinant parvovirus. Cancer Gene Ther 2010; 18:240-9. [PMID: 21102423 DOI: 10.1038/cgt.2010.73] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Preclinical studies using various cell culture and animal systems highlight the potential of recombinant rodent parvoviruses (recPVs) for cancer therapy. Production of these viruses is, however, not efficient and this hampers the clinical applications of these agents. In this study, we show that the adenovirus genes E2a, E4(orf6) and VA RNA increase the production of recPVs by more than 10-fold and reduce the time of production from 3 to 2 days in HEK293T cells. The helper effects of these genes can be observed with different recPVs, regardless of the nature and size of the inserted transgene. Furthermore, we generated a recombinant Adenovirus 5 carrying the parvovirus VP transcription unit. This helper, named Ad-VP, allows recPVs to be efficiently produced through a protocol based only on cell infection, making possible to use cell lines, such as NB324K, which are good producers of parvoviruses but are hardly transfectable. Hence, we could further improve viral titers and reduce time and costs of production. This Ad-VP helper-based protocol could be scaled up to a bioreactor format for the generation of the large amounts of recPVs needed for future clinical applications.
Collapse
|
32
|
Janus LM, Smoczek A, Jörns A, Hedrich HJ, Bleich A. Presence of Minute virus of mice in immunocompetent mice despite the onset of host immunity. Vet Microbiol 2010; 146:51-8. [DOI: 10.1016/j.vetmic.2010.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 04/13/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
|
33
|
Dempe S, Stroh-Dege AY, Schwarz E, Rommelaere J, Dinsart C. SMAD4: a predictive marker of PDAC cell permissiveness for oncolytic infection with parvovirus H-1PV. Int J Cancer 2010; 126:2914-27. [PMID: 19856310 DOI: 10.1002/ijc.24992] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the eighth frequent solid tumor and fourth leading cause of cancer death. Because current treatments against PDAC are still unsatisfactory, new anticancer strategies are required, including oncolytic viruses. Among these, autonomous parvoviruses (PV), like MVMp (minute virus of mice) and H-1PV are being explored as candidates for cancer gene therapy. Human PDAC cell lines were identified to display various susceptibilities to an infection with H-1PV. The correlation between the integrity of the transcription factor SMAD4, mutated in 50% of all PDAC, and H-1PV permissiveness was particularly striking. Indeed, mutation or deletion of SMAD4 dramatically reduced the activity of the P4 promoter and, consequently, the accumulation of the pivotal NS1 protein. By means of DNA affinity immunoblotting, novel binding sites for SMAD4 and c-JUN transcription factors could be identified in the P4 promoter of H-1PV. The overexpression of wild-type SMAD4 in deficient cell lines (AsPC-1, Capan-1) stimulated the activity of the P4 promoter, whereas interference of endogenous SMAD4 function with a dominant-negative mutant decreased the viral promoter activity in wild-type SMAD4-expressing cells (Panc-1, MiaPaCa-2) reducing progeny virus production. In conclusion, the importance of members of the SMAD family for H-1PV early promoter P4 activity should guide us to select SMAD4-positive PDACs, which may be possible targets for an H-1PV-based cancer therapy.
Collapse
Affiliation(s)
- Sebastian Dempe
- Abt F010, Infection and Cancer Program, Tumor Virology Division, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 242, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
34
|
Wizla P, Begue A, Loison I, Richard A, Caillet-Fauquet P, Stéhelin D. Ectopic expression of H-1 parvovirus NS1 protein induces alterations in actin filaments and cell death in human normal MRC-5 and transformed MRC-5 SV2 cells. Arch Virol 2010; 155:771-5. [PMID: 20237811 DOI: 10.1007/s00705-010-0637-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 02/09/2010] [Indexed: 11/28/2022]
Abstract
When grown in human cell lines, oncolytic H-1 parvovirus (H-1PV) replication preferentially occurs in transformed cells, which ultimately die upon infection. H-1PV-induced cytotoxicity is mainly due to P4 promoter-driven NS1 protein expression. Infection of untransformed cells generally does not induce deleterious effects because the P4 promoter is not activated. Here, we show that ectopic CMV-driven NS1 protein expression in normal human MRC-5 cells results in alterations of actin filaments and cell death, and both effects are prevented by a serine 473 mutation. The same substitution preserves actin filaments of transfected MRC-5 SV2 cells, that are MRC-5 transformed counterparts, but does not impair NS1-induced cytotoxicity.
Collapse
Affiliation(s)
- Pierre Wizla
- UMR 8161, CNRS, Institut de Biologie de Lille, 1 rue du Professeur Calmette, BP 447, 59021, Lille Cedex, France.
| | | | | | | | | | | |
Collapse
|
35
|
Activation of an antiviral response in normal but not transformed mouse cells: a new determinant of minute virus of mice oncotropism. J Virol 2010; 84:516-31. [PMID: 19864388 DOI: 10.1128/jvi.01618-09] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Parvovirus minute virus of mice (MVMp) is endowed with oncotropic properties so far ascribed only to the dependency of the virus life cycle on cellular factors expressed during S phase and/or modulated by malignant transformation. For other viruses oncotropism relies on their inability to circumvent type I interferon (IFN)-induced innate antiviral mechanisms, the first line of defense triggered by normal cells against viral infections. These agents propagate, therefore, preferentially in transformed/tumor cells, which often lack functional antiviral mechanisms. The present study aimed at investigating whether antiviral processes also contribute to MVMp oncotropism. Our results demonstrate that in contrast to MVMp-permissive transformed mouse A9 fibroblasts, freshly isolated normal counterparts (mouse embryonic fibroblasts [MEFs]) mount, through production and release of type I IFNs upon their infection, an antiviral response against MVMp lytic multiplication. Pretreatment of MEFs with a type I IFN-beta-neutralizing antibody, prior to MVMp infection, inhibits the virus-triggered antiviral response and improves the fulfillment of the MVMp life cycle. Our results also show that part of the A9 permissiveness to MVMp relies on the inability to produce type I IFNs upon parvovirus infection, a feature related either to an A9 intrinsic deficiency of this process or to an MVMp-triggered inhibitory mechanism, since stimulation of these cells by exogenous IFN-beta strongly inhibits the parvovirus life cycle. Taken together, our results demonstrate for the first time that parvovirus infection triggers an innate antiviral response in normal cells and suggest that the MVMp oncotropism depends at least in part on the failure of infected transformed cells to mount such a response.
Collapse
|
36
|
Angelova AL, Aprahamian M, Grekova SP, Hajri A, Leuchs B, Giese NA, Dinsart C, Herrmann A, Balboni G, Rommelaere J, Raykov Z. Improvement of gemcitabine-based therapy of pancreatic carcinoma by means of oncolytic parvovirus H-1PV. Clin Cancer Res 2009; 15:511-9. [PMID: 19147756 DOI: 10.1158/1078-0432.ccr-08-1088] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
UNLABELLED Pancreatic carcinoma is a gastrointestinal malignancy with poor prognosis. Treatment with gemcitabine, the most potent chemotherapeutic against this cancer up to date, is not curative, and resistance may appear. Complementary treatment with an oncolytic virus, such as the rat parvovirus H-1PV, which is infectious but nonpathogenic in humans, emerges as an innovative option. PURPOSE To prove that combining gemcitabine and H-1PV in a model of pancreatic carcinoma may reduce the dosage of the toxic drug and/or improve the overall anticancer effect. EXPERIMENTAL DESIGN Pancreatic tumors were implanted orthotopically in Lewis rats or subcutaneously in nude mice and treated with gemcitabine, H-1PV, or both according to different regimens. Tumor size was monitored by micro-computed tomography, whereas bone marrow, liver, and kidney functions were monitored by measuring clinically relevant markers. Human pancreatic cell lines and gemcitabine-resistant derivatives were tested in vitro for sensitivity to H-1PV infection with or without gemcitabine. RESULTS In vitro studies proved that combining gemcitabine with H-1PV resulted in synergistic cytotoxic effects and achieved an up to 15-fold reduction in the 50% effective concentration of the drug, with drug-resistant cells remaining sensitive to virus killing. Toxicologic screening showed that H-1PV had an excellent safety profile when applied alone or in combination with gemcitabine. The benefits of applying H-1PV as a second-line treatment after gemcitabine included reduction of tumor growth, prolonged survival of the animals, and absence of metastases on CT-scans. CONCLUSION In addition to their potential use as monotherapy for pancreatic cancer, parvoviruses can be best combined with gemcitabine in a two-step protocol.
Collapse
Affiliation(s)
- Assia L Angelova
- Infection and Cancer Programme F010 and INSERM U701, German Cancer Research Center, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Oncolytic rat parvovirus H-1PV, a candidate for the treatment of human lymphoma: In vitro and in vivo studies. Mol Ther 2009; 17:1164-72. [PMID: 19367260 DOI: 10.1038/mt.2009.78] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The incidence of lymphomas developing in both immunocompetent and immunosuppressed patients continues to steadily increase worldwide. Current chemotherapy and immunotherapy approaches have several limitations, such as severe side toxicity and selection of resistant cell variants. Autonomous parvoviruses (PVs), in particular the rat parvovirus H-1PV, have emerged as promising anticancer agents. Although it is apathogenic in humans, H-1PV has been shown to infect and suppress various rat and human tumors in animal models. In this study, we demonstrate the capacity of H-1PV for efficiently killing, through necrosis, cell cultures originating from Burkitt's lymphoma (BL), while sparing normal B lymphocytes. The cytotoxic effect was generally accompanied by a productive H-1PV infection. Remarkably, parvovirus-based monotherapy efficiently suppressed established BL at an advanced stage in a severe combined immunodeficient (SCID) mouse model of the disease. The data show for the first time that an oncolytic parvovirus deserves further consideration as a potential tool for the treatment of some non-Hodgkin B-cell lymphomas, including those resistant to apoptosis induction by rituximab.
Collapse
|
38
|
Abstract
Advances in genetics, proteomics and cellular and molecular biology are being integrated and translated to develop effective methods for the prevention and control of cancer. One such combined effort is to create multifunctional nanodevices that will specifically recognize tumors and thus enable early diagnosis and provide targeted treatment of this disease. Viral particles are being considered for this purpose since they are inherently nanostructures with well-defined geometry and uniformity, ideal for displaying molecules in a precise spatial distribution at the nanoscale level and subject to greater structural control. Viruses are presumably the most efficient nanocontainer for cellular delivery as they have naturally evolved mechanisms for binding to and entering cells. Virus-based systems typically require genetic or chemical modification of their surfaces to achieve tumor-specific interactions. Interestingly, canine parvovirus (CPV) has a natural affinity for transferrin receptors (TfRs) (both of canine and human origin) and this property could be harnessed as TfRs are overexpressed by a variety of human tumor cells. Since TfR recognition relies on the CPV capsid protein, we envisioned the use of virus or its shells as tumor targeting agents. We observed that derivatization of CPV virus-like particles (VLPs) with dye molecules did not impair particle binding to TfRs or internalization into human tumor cells. Thus CPV-based VLPs with a natural tropism for TfRs hold great promise in the development of novel nanomaterial for delivery of a therapeutic and/or genetic cargo.
Collapse
Affiliation(s)
- P Singh
- Division of Hematology and Oncology, Department of Medicine, Building 23, Room 436A, UCI Medical Center, 101 City Drive South, Orange, CA 92868, USA.
| |
Collapse
|
39
|
Enderlin M, Kleinmann EV, Struyf S, Buracchi C, Vecchi A, Kinscherf R, Kiessling F, Paschek S, Sozzani S, Rommelaere J, Cornelis JJ, Van Damme J, Dinsart C. TNF-alpha and the IFN-gamma-inducible protein 10 (IP-10/CXCL-10) delivered by parvoviral vectors act in synergy to induce antitumor effects in mouse glioblastoma. Cancer Gene Ther 2008; 16:149-60. [PMID: 18670452 DOI: 10.1038/cgt.2008.62] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Interferon-gamma-inducible protein 10 is a potent chemoattractant for natural killer cells and activated T lymphocytes. It also displays angiostatic properties and some antitumor activity. Tumor necrosis factor-alpha (TNF-alpha) is a powerful immunomodulating cytokine with demonstrated tumoricidal activity in various tumor models and the ability to induce strong immune responses. This prompted us to evaluate the antitumor effects of recombinant parvoviruses designed to deliver IP-10 or TNF-alpha into a glioblastoma. When Gl261 murine glioma cells were infected in vitro with an IP-10- or TNF-alpha-transducing parvoviral vector and were subcutaneously implanted in mice, tumor growth was significantly delayed. Complete tumor regression was observed when the glioma cells were coinfected with both the vectors, demonstrating synergistic antitumor activity. In an established in vivo glioma model, however, repeated simultaneous peritumoral injection of the IP-10- and TNF-alpha-delivering parvoviruses failed to improve the therapeutic effect as compared with the use of a single cytokine-delivering vector. In this tumor model, cytokine-mediated immunostimulation, rather than inhibition of vascularization, is likely responsible for the therapeutic efficacy.
Collapse
Affiliation(s)
- M Enderlin
- Deutsches Krebsforschungszentrum, Infection and Cancer Program, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sieben M, Herzer K, Zeidler M, Heinrichs V, Leuchs B, Schuler M, Cornelis JJ, Galle PR, Rommelaere J, Moehler M. Killing of p53-deficient hepatoma cells by parvovirus H-1 and chemotherapeutics requires promyelocytic leukemia protein. World J Gastroenterol 2008; 14:3819-28. [PMID: 18609705 PMCID: PMC2721438 DOI: 10.3748/wjg.14.3819] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the synergistic targeting and killing of human hepatocellular carcinoma (HCC) cells lacking p53 by the oncolytic autonomous parvovirus (PV) H-1 and chemotherapeutic agents and its dependence on functional promyelocytic leukemia protein (PML).
METHODS: The role of p53 and PML in regulating cytotoxicity and gene transfer mediated by wild-type (wt) PV H-1 were explored in two pairs of isogenic human hepatoma cell lines with different p53 status. Furthermore, H-1 PV infection was combined with cytostatic drug treatment.
RESULTS: While the HCC cells with different p53 status studied were all susceptible to H-1 PV-induced apoptosis, the cytotoxicity of H-1 PV was more pronounced in p53-negative than in p53-positive cells. Apoptosis rates in p53-negative cell lines treated by genotoxic drugs were further enhanced by a treatment with H-1 PV. In flow cytometric analyses, H-1 PV infection resulted in a reduction of the mitochondrial transmembrane potential. In addition, H-1 PV cells showed a significant increase in PML expression. Knocking down PML expression resulted in a striking reduction of the level of H-1 PV infected tumor cell death.
CONCLUSION: H-1 PV is a suitable agent to circumvent the resistance of p53-negative HCC cells to genotoxic agents, and it enhances the apoptotic process which is dependent on functional PML. Thus, H-1 PV and its oncolytic vector derivatives may be considered as therapeutic options for HCC, particularly for p53-negative tumors.
Collapse
|
41
|
Raykov Z, Rommelaere J. Potential of tumour cells for delivering oncolytic viruses. Gene Ther 2008; 15:704-10. [DOI: 10.1038/gt.2008.34] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
Kirn DH. Monthly Update: Oncologic, Endocrine & Metabolic: Replicating oncolytic viruses: An overview. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.5.6.753] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- David H Kirn
- Onyx Pharmaceuticals, Richmond, California & University of California, San Francisco, California, USA
| |
Collapse
|
43
|
Wang JH, Zhang WP, Liu HX, Wang D, Li YF, Wang WQ, Wang L, He FR, Wang Z, Yan QG, Chen LW, Huang GS. Detection of human parvovirus B19 in papillary thyroid carcinoma. Br J Cancer 2008; 98:611-8. [PMID: 18212749 PMCID: PMC2243166 DOI: 10.1038/sj.bjc.6604196] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
To evaluate whether parvovirus B19, a common human pathogen, was also involved in papillary thyroid carcinoma (PTC), 112 paraffin-embedded thyroid specimens of benign nodules, papillary, medullary and follicular carcinomas, and normal controls were examined for B19 DNA and capsid protein by nested PCR, in situ hybridisation (ISH) and immunohistochemistry (IHC). The expression of the nuclear factor-κB (NF-κB) was investigated by IHC. The results showed B19 DNA commonly exists in human thyroid tissues; however, there were significant differences between PTC group and normal controls, and between PTC and nonneoplastic adjacent tissues (P<0.001). The presence of viral DNA in PTC neoplastic epithelium was confirmed by laser-capture microdissection and sequencing of nested PCR products. B19 capsid protein in PTC group was significantly higher than that of all the control groups and nonneoplastic adjacent tissues (P⩽0.001). Compared with control groups, the activation of NF-κB in PTC group was significantly increased (P⩽0.02), except for medullary carcinomas, and the activation of NF-κB was correlated with the viral protein presence (P=0.002). Moreover, NF-κB was colocalised with B19 DNA in the neoplastic epithelium of PTC by double staining of IHC and ISH. These results indicate for the first time a possible role of B19 in pathogenesis of PTC.
Collapse
Affiliation(s)
- J H Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Augmented transgene expression in transformed cells using a parvoviral hybrid vector. Cancer Gene Ther 2008; 15:252-67. [PMID: 18202715 DOI: 10.1038/sj.cgt.7701113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Autonomous parvoviruses possess an intrinsic oncotropism based on viral genetic elements controlling gene expression and genome replication. We constructed a hybrid vector consisting of the H1 parvovirus-derived expression cassette comprising the p4 promoter, the ns1 gene and the p38 promoter flanked by the adeno-associated viruses 2 (AAV2) inverted terminal repeats and packaged into AAV2 capsids. Gene transduction using this vector could be stimulated by coinfection with adenovirus, by irradiation or treatment with genotoxic agents, similar to standard AAV2 vectors. However, the latter were in most cases less efficient in gene transduction than the hybrid vector. With the new vector, tumor cell-selective increase in transgene expression was observed in pairs of transformed and non-transformed cells, leading to selective killing of the transformed cells after expression of a prodrug-converting enzyme. Preferential gene expression in tumor versus normal liver tissue was also observed in vivo in a syngeneic rat model. Comparative transduction of a panel of different tumor cell lines with the H1 and the H1/AAV hybrid vector showed a preference of each vector for distinct cell types, probably reflecting the dependence of the viral tropism on capsid determinants.
Collapse
|
45
|
Wetzel K, Struyf S, Van Damme J, Kayser T, Vecchi A, Sozzani S, Rommelaere J, Cornelis JJ, Dinsart C. MCP-3 (CCL7) delivered by parvovirus MVMp reduces tumorigenicity of mouse melanoma cells through activation of T lymphocytes and NK cells. Int J Cancer 2007; 120:1364-71. [PMID: 17154174 DOI: 10.1002/ijc.22421] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Monocyte chemotactic protein 3 (MCP-3/CCL7), a CC chemokine able to attract and activate a large panel of leukocytes including natural killer cells and T lymphocytes, could be beneficial in antitumor therapy. Vectors were constructed based on the autonomous parvovirus minute virus of mice (MVMp), carrying the human (MCP-3) cDNA. These vectors were subsequently evaluated in the poorly immunogenic mouse melanoma model B78/H1. The infection of the tumor cells with MCP3-transducing vector at low virus input multiplicities, but not with wild-type virus, strongly inhibited tumor growth after implantation in euthymic mice. In a therapeutic B78/H1 model, repeated intratumoral injections of MCP3-tranducing virus prevented further tumor expansion as long as the treatment was pursued. The antitumor effects of the MCP-3-transducing vector were not restricted to this tumor model since they could also be observed in the K1735 melanoma. The depletion of CD4, CD8, NK cells and of interferon gamma (IFNgamma) in mice implanted with MVMp/MCP3-infected B78/H1 cells abolished the antitumor activity of the vector. The latter data, together with tumor growth in nude mice and reverse-transcriptase (RT)-PCR analyses of MVMp/MCP3-treated tumors, clearly showed that activated CD4, CD8 and NK cells were indispensable for the antineoplastic effect in the B78/H1 tumor. Altogether, our results show that MCP3-transducing parvovirus vectors may be quite potent against poorly or nonimmunogenic tumors, even in conditions where only a fraction of the tumor cell population is efficiently infected with recombinant parvoviruses.
Collapse
Affiliation(s)
- Kristiane Wetzel
- Infection and Cancer Program, Abteilung F010, and Institut National de la Santé et de la Recherche Médicale U701, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Di Piazza M, Mader C, Geletneky K, Herrero Y Calle M, Weber E, Schlehofer J, Deleu L, Rommelaere J. Cytosolic activation of cathepsins mediates parvovirus H-1-induced killing of cisplatin and TRAIL-resistant glioma cells. J Virol 2007; 81:4186-98. [PMID: 17287256 PMCID: PMC1866092 DOI: 10.1128/jvi.02601-06] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gliomas are often resistant to the induction of apoptotic cell death as a result of the development of survival mechanisms during astrocyte malignant transformation. In particular, the overexpression of Bcl-2-family members interferes with apoptosis initiation by DNA-damaging agents (e.g., cisplatin) or soluble death ligands (e.g., TRAIL). Using low-passage-number cultures of glioma cells, we have shown that parvovirus H-1 is able to induce death in cells resistant to TRAIL, cisplatin, or both, even when Bcl-2 is overexpressed. Parvovirus H-1 triggers cell death through both the accumulation of lysosomal cathepsins B and L in the cytosol of infected cells and the reduction of the levels of cystatin B and C, two cathepsin inhibitors. The impairment of either of these effects protects glioma cells from the viral lytic effect. In normal human astrocytes, parvovirus H-1 fails to induce a killing mechanism. In vivo, parvovirus H-1 infection of rat glioma cells intracranially implanted into recipient animals triggers cathepsin B activation as well. This report identifies for the first time cellular effectors of the killing activity of parvovirus H-1 against malignant brain cells and opens up a therapeutic approach which circumvents their frequent resistance to other death inducers.
Collapse
Affiliation(s)
- Matteo Di Piazza
- Infection and Cancer Program, Division F010 and INSERM Unit 701, and German Cancer Research Center, Division F010, Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Paglino J, Burnett E, Tattersall P. Exploring the contribution of distal P4 promoter elements to the oncoselectivity of Minute Virus of Mice. Virology 2006; 361:174-84. [PMID: 17175002 PMCID: PMC1853334 DOI: 10.1016/j.virol.2006.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2006] [Revised: 10/13/2006] [Accepted: 11/03/2006] [Indexed: 11/20/2022]
Abstract
Minute Virus of Mice (MVM) shares inherent oncotropic properties with other members of the genus Parvovirus. Two elements responsible, at least in part, for this oncoselectivity have been mapped to an Ets1 binding site adjacent to the P4 TATA box of the initiating promoter, P4, and to a more distal cyclic AMP responsive element (CRE), located within the telomeric hairpin stem. Here the CRE overlaps one half-site for the binding of parvoviral initiation factor (PIF), which is essential for viral DNA replication. We used a degenerate oligonucleotide selection approach to show that CRE binding protein (CREB) selects the sequence ACGTCAC within this context, rather than its more generally accepted palindromic TGACGTCA recognition site. We have developed strategies for manipulating these sequences directly within the left-end palindrome of the MVM infectious clone and used them to clone mutants whose CRE either matches the symmetric consensus sequence or is scrambled, or in which the PIF binding site is incrementally weakened with respect to the CRE. The panel of mutants were tested for fitness relative to wildtype in normal murine fibroblasts A9 or transformed human fibroblasts 324 K, through multiple rounds of growth in co-infected cultures, using a differential real-time quantitative PCR assay. We confirmed that inactivating the CRE substantially abrogates oncoselectivity, but found that improving its fit to the palindromic consensus is somewhat debilitating in either cell type. We also confirmed that reducing the PIF half-site spacing by one basepair enhances oncoselectivity, but found that a further basepair deletion significantly reduces this effect.
Collapse
Affiliation(s)
- Justin Paglino
- Department of Laboratory Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
- Graduate Program in Investigative Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
| | - Erik Burnett
- Department of Laboratory Genetics, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
| | - Peter Tattersall
- Department of Laboratory Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
- Department of Laboratory Genetics, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA
- * Corresponding author. Department of Laboratory Medicine, Yale University Medical School, 333 Cedar Street, New Haven, CT 067510, USA. Fax: +1 203 688 7340. E-mail address: (P. Tattersall)
| |
Collapse
|
48
|
Post A. Environmental exposure to bacteria and viruses may provide oncolytic protection against cancers, and declining exposure to infections may contribute to a rising incidence of cancer. Med Hypotheses 2006; 68:558-61. [PMID: 17030097 DOI: 10.1016/j.mehy.2006.02.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 02/26/2006] [Accepted: 02/28/2006] [Indexed: 10/24/2022]
Abstract
Over the past century, the incidence of cancer rose markedly in developed countries. Many underlying factors are known, but not all of them. Over the same century, advances in public health and hygiene practically eradicated many infectious illnesses, and exposure to some untargeted infectious agents has also declined. There are reasons to suspect that these two trends are linked. Although some viruses and one bacterium are known to be oncogenic, a high percentage of infectious agents, 71% of viruses tested in one study, have shown oncolytic properties. Some of these agents have minimal or no clinical consequences, and some are - or were - quite ubiquitous. Many types of cancer are unusually susceptible to several different agents, sometimes 10-1000 times more susceptible than normal cells. When wild-type or attenuated viruses are used to treat cancer, they are often more effective with smaller tumors, and it may be that nascent, undetectable cancers are especially vulnerable. Environmental exposure to bacteria and viruses may provide natural protection from cancer by attacking it at this stage, or inducing the body to do so. Immunity to an infectious agent can attenuate its oncolytic effect, but generally does not stop it, so both initial and repeated exposures may be protective. As more direct evidence, treatment of animals with selected live virus vaccines has provided protection from cancer, and in humans, exposure to two specific infectious agents is known to correlate with a reduced risk of cancer. The significant decline in exposure to infectious agents over the past century may have inadvertently weakened this naturally protective mechanism, driving cancer rates up. There is considerable research on the impact of oncogenic agents, but surprisingly little regarding the impact of oncolytic agents on cancer rates. An understanding of the relationship between natural infections and the suppression of cancer may lead to prophylactic measures against cancer, improved public health policies regarding vaccination programs and hygiene, and greater insight into cancer treatment as well.
Collapse
Affiliation(s)
- A Post
- Department of Mechanical and Manufacturing Engineering Technology, Arizona State University, 7441 E. Tillman, Mesa, AZ 85212, USA.
| |
Collapse
|
49
|
Servais C, Caillet-Fauquet P, Draps ML, Velu T, de Launoit Y, Brandenburger A. Hypoxic-response elements in the oncolytic parvovirus Minute virus of mice do not allow for increased vector production at low oxygen concentration. J Gen Virol 2006; 87:1197-1201. [PMID: 16603521 DOI: 10.1099/vir.0.81754-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Vectors derived from the autonomous parvovirus Minute virus of mice, MVM(p), are promising tools for the gene therapy of cancer. The validation of their in vivo anti-tumour effect is, however, hampered by the difficulty to produce high-titre stocks. In an attempt to increase vector titres, host cells were subjected to low oxygen tension (hypoxia). It has been shown that a number of viruses are produced at higher titres under these conditions. This is the case, among others, for another member of the family Parvoviridae, the erythrovirus B19 virus. Hypoxia stabilizes a hypoxia-inducible transcription factor (HIF-1alpha) that interacts with a 'hypoxia-responsive element' (HRE), the consensus sequence of which ((A)/(G)CGTG) is present in the B19 and MVM promoters. Whilst the native P4 promoter was induced weakly in hypoxia, vector production was reduced dramatically, and adding HRE elements to the P4 promoter of the vector did not alleviate this reduction. Hypoxia has many effects on cell metabolism. Therefore, even if the P4 promoter is activated, the cellular factors that are required for the completion of the parvoviral life cycle may not be expressed.
Collapse
Affiliation(s)
- Charlotte Servais
- Laboratoire de Cytologie et de Cancérologie Expérimentale, IBMM-IRIBHM, Université Libre de Bruxelles, 12 rue des Professeurs Jeener et Brachet, B-6041 Gosselies, Belgium
| | - Perrine Caillet-Fauquet
- Laboratoire de Virologie Moléculaire, Faculté de Médecine, Campus Erasme, Université Libre de Bruxelles, Route de Lennik 808, B-1070 Bruxelles, Belgium
| | - Marie-Louise Draps
- Laboratoire de Virologie Moléculaire, Faculté de Médecine, Campus Erasme, Université Libre de Bruxelles, Route de Lennik 808, B-1070 Bruxelles, Belgium
| | - Thierry Velu
- Laboratoire de Cytologie et de Cancérologie Expérimentale, IBMM-IRIBHM, Université Libre de Bruxelles, 12 rue des Professeurs Jeener et Brachet, B-6041 Gosselies, Belgium
| | - Yvan de Launoit
- UMR 8117 CNRS, Université de Lille 1, Institut Pasteur Lille, Institut de Biologie de Lille, Lille, France
- Laboratoire de Virologie Moléculaire, Faculté de Médecine, Campus Erasme, Université Libre de Bruxelles, Route de Lennik 808, B-1070 Bruxelles, Belgium
| | - Annick Brandenburger
- Laboratoire de Cytologie et de Cancérologie Expérimentale, IBMM-IRIBHM, Université Libre de Bruxelles, 12 rue des Professeurs Jeener et Brachet, B-6041 Gosselies, Belgium
| |
Collapse
|
50
|
Malerba M, Nikolova D, Cornelis J, Iggo R. Targeting of autonomous parvoviruses to colon cancer by insertion of Tcf sites in the P4 promoter. Cancer Gene Ther 2006; 13:273-80. [PMID: 16151476 DOI: 10.1038/sj.cgt.7700904] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Wnt signaling pathway is activated by mutations in the adenomatous polyposis coli (APC) or beta-catenin genes in most colon cancers, leading to the transactivation of promoters containing binding sites for the Tcf/LEF family of transcription factors. We have previously shown that it is possible to confer colon cancer specificity on autonomous parvoviruses by inserting Tcf sites into the viral P4 promoter. The mutant Tcf promoters were responsive to activation of the Wnt pathway but the viruses replicated poorly. We show here that reduction of the number of Tcf sites from four to two leads to an increase in the efficiency of replication and toxicity of the viruses in Co115 colon cancer cells, with only a small reduction in selectivity for cells with an active Wnt signaling pathway. Despite this improvement, virus production by most colon cancer cells remained low. Analysis of parental phH1 virus infection of SW480 colon cancer cells showed that the nonstructural and capsid proteins were expressed, but single stranded DNA and progeny virus were not produced. This defect reflects the dependence of autonomous parvoviruses on host functions for many steps in their replication cycle and represents a major limitation to the use of selectively replicating parvoviruses for colon cancer therapy.
Collapse
Affiliation(s)
- M Malerba
- NCCR Molecular Oncology, Swiss Institute for Experimental Cancer Research, ISREC, Epalinges, Switzerland
| | | | | | | |
Collapse
|