1
|
Zhu Z, McGray AJR, Jiang W, Lu B, Kalinski P, Guo ZS. Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer 2022; 21:196. [PMID: 36221123 PMCID: PMC9554963 DOI: 10.1186/s12943-022-01664-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
Oncolytic viruses (OVs) represent a new class of multi-modal immunotherapies for cancer, with OV-elicited antitumor immunity being key to their overall therapeutic efficacy. Currently, the clinical effectiveness of OV as monotherapy remains limited, and thus investigators have been exploring various combinations with other anti-cancer agents and demonstrated improved therapeutic efficacy. As cancer cells have evolved to alter key signaling pathways for enhanced cell proliferation, cancer progression and metastasis, these cellular and molecular changes offer promising targets for rational cancer therapy design. In this regard, key molecules in relevant signaling pathways for cancer cells or/and immune cells, such as EGFR-KRAS (e.g., KRASG12C), PI3K-AKT-mTOR, ERK-MEK, JAK-STAT, p53, PD-1-PD-L1, and epigenetic, or immune pathways (e.g., histone deacetylases, cGAS-STING) are currently under investigation and have the potential to synergize with OV to modulate the immune milieu of the tumor microenvironment (TME), thereby improving and sustaining antitumor immunity. As many small molecule modulators of these signaling pathways have been developed and have shown strong therapeutic potential, here we review key findings related to both OV-mediated immunotherapy and the utility of small molecule modulators of signaling pathways in immuno-oncology. Then, we focus on discussion of the rationales and potential strategies for combining OV with selected modulators targeting key cellular signaling pathways in cancer or/and immune cells to modulate the TME and enhance antitumor immunity and therapeutic efficacy. Finally, we provide perspectives and viewpoints on the application of novel experimental systems and technologies that can propel this exciting branch of medicine into a bright future.
Collapse
Affiliation(s)
- Zhi Zhu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - A J Robert McGray
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Weijian Jiang
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Binfeng Lu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | - Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
2
|
Sunil AA, Skaria T. Novel regulators of airway epithelial barrier function during inflammation: potential targets for drug repurposing. Expert Opin Ther Targets 2022; 26:119-132. [PMID: 35085478 DOI: 10.1080/14728222.2022.2035720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Endogenous inflammatory signaling molecules resulting from deregulated immune responses, can impair airway epithelial barrier function and predispose individuals with airway inflammatory diseases to exacerbations and lung infections. Targeting the specific endogenous factors disrupting the airway barrier therefore has the potential to prevent disease exacerbations without affecting the protective immune responses. AREAS COVERED Here, we review the endogenous factors and specific mechanisms disrupting airway epithelial barrier during inflammation and reflect on whether these factors can be specifically targeted by repurposed existing drugs. Literature search was conducted using PubMed, drug database of US FDA and European Medicines Agency until and including September 2021. EXPERT OPINION IL-4 and IL-13 signaling are the major pathways disrupting the airway epithelial barrier during airway inflammation. However, blocking IL-4/IL-13 signaling may adversely affect protective immune responses and increase susceptibility of host to infections. An alternate approach to modulate airway epithelial barrier function involves targeting specific downstream component of IL-4/IL-13 signaling or different inflammatory mediators responsible for regulation of airway epithelial barrier. Airway epithelium-targeted therapy using inhibitors of HDAC, HSP90, MIF, mTOR, IL-17A and VEGF may be a potential strategy to prevent airway epithelial barrier dysfunction in airway inflammatory diseases.
Collapse
Affiliation(s)
- Ahsan Anjoom Sunil
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Tom Skaria
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
3
|
Gilchrist VH, Jémus-Gonzalez E, Said A, Alain T. Kinase inhibitors with viral oncolysis: Unmasking pharmacoviral approaches for cancer therapy. Cytokine Growth Factor Rev 2020; 56:83-93. [PMID: 32690442 DOI: 10.1016/j.cytogfr.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/28/2022]
Abstract
There are more than 500 kinases in the human genome, many of which are oncogenic once constitutively activated. Fortunately, numerous hyperactive kinases are druggable, and several targeted small molecule kinase inhibitors have demonstrated impressive clinical benefits in cancer treatment. However, their often cytostatic rather than cytotoxic effect on cancer cells, and the development of resistance mechanisms, remain significant limitations to these targeted therapies. Oncolytic viruses are an emerging class of immunotherapeutic agents with a specific oncotropic nature and excellent safety profile, highlighting them as a promising alternative to conventional therapeutic modalities. Nonetheless, the clinical efficacy of oncolytic virotherapy is challenged by immunological and physical barriers that limit viral delivery, replication, and spread within tumours. Several of these barriers are often associated with oncogenic kinase activity and, in some cases, worsened by the action of oncolytic viruses on kinase signaling during infection. What if inhibiting these kinases could potentiate the cancer-lytic and anti-tumour immune stimulating properties of oncolytic virotherapies? This could represent a paradigm shift in the use of specific kinase inhibitors in the clinic and provide a novel therapeutic approach to the treatment of cancers. A phase III clinical trial combining the oncolytic Vaccinia virus Pexa-Vec with the kinase inhibitor Sorafenib was initiated. While this trial failed to show any benefits over Sorafenib monotherapy in patients with advanced liver cancer, several pre-clinical studies demonstrate that targeting kinases combined with oncolytic viruses have synergistic effects highlighting this strategy as a unique avenue to cancer therapy. Herein, we review the combinations of oncolytic viruses with kinase inhibitors reported in the literature and discuss the clinical opportunities that represent these pharmacoviral approaches.
Collapse
Affiliation(s)
- Victoria Heather Gilchrist
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| | - Estephanie Jémus-Gonzalez
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada
| | - Aida Said
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
4
|
Brachtlova T, van Beusechem VW. Unleashing the Full Potential of Oncolytic Adenoviruses against Cancer by Applying RNA Interference: The Force Awakens. Cells 2018; 7:cells7120228. [PMID: 30477117 PMCID: PMC6315459 DOI: 10.3390/cells7120228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/23/2022] Open
Abstract
Oncolytic virus therapy of cancer is an actively pursued field of research. Viruses that were once considered as pathogens threatening the wellbeing of humans and animals alike are with every passing decade more prominently regarded as vehicles for genetic and oncolytic therapies. Oncolytic viruses kill cancer cells, sparing healthy tissues, and provoke an anticancer immune response. Among these viruses, recombinant adenoviruses are particularly attractive agents for oncolytic immunotherapy of cancer. Different approaches are currently examined to maximize their therapeutic effect. Here, knowledge of virus–host interactions may lead the way. In this regard, viral and host microRNAs are of particular interest. In addition, cellular factors inhibiting viral replication or dampening immune responses are being discovered. Therefore, applying RNA interference is an attractive approach to strengthen the anticancer efficacy of oncolytic viruses gaining attention in recent years. RNA interference can be used to fortify the virus’ cancer cell-killing and immune-stimulating properties and to suppress cellular pathways to cripple the tumor. In this review, we discuss different ways of how RNA interference may be utilized to increase the efficacy of oncolytic adenoviruses, to reveal their full potential.
Collapse
Affiliation(s)
- Tereza Brachtlova
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, De Boelelaan 1117, 1007 MB Amsterdam, The Netherlands.
| | - Victor W van Beusechem
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, De Boelelaan 1117, 1007 MB Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Phan M, Watson MF, Alain T, Diallo JS. Oncolytic Viruses on Drugs: Achieving Higher Therapeutic Efficacy. ACS Infect Dis 2018; 4:1448-1467. [PMID: 30152676 DOI: 10.1021/acsinfecdis.8b00144] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Over the past 20 years there has been a dramatic expansion in the testing of oncolytic viruses (OVs) for the treatment of cancer. OVs are unique biotherapeutics that induce multimodal responses toward tumors, from direct cytopathic effects on cancer cells, to tumor associated blood vessel disruption, and ultimately potent stimulation of anti-tumor immune activation. These agents are highly targeted and can be efficacious as cancer treatments resulting in some patients experiencing complete tumor regression and even cures from OV monotherapy. However, most patients have limited responses with viral replication in tumors often found to be modest and transient. To augment OV replication, increase bystander killing of cancer cells, and/or stimulate stronger targeted anti-cancer immune responses, drug combination approaches have taken center stage for translation to the clinic. Here we comprehensively review drugs that have been combined with OVs to increase therapeutic efficacy, examining the proposed mechanisms of action, and we discuss trends in pharmaco-viral immunotherapeutic approaches currently being investigated.
Collapse
Affiliation(s)
- Michael Phan
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Margaret F. Watson
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Children’s Hospital of Eastern Ontario Research Institute, 401 Smyth Road Research Building 2, Second Floor, Room 2119, Ottawa, Ontario K1H 8L1, Canada
| | - Tommy Alain
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Children’s Hospital of Eastern Ontario Research Institute, 401 Smyth Road Research Building 2, Second Floor, Room 2119, Ottawa, Ontario K1H 8L1, Canada
| | - Jean-Simon Diallo
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
6
|
Comins C, Simpson GR, Rogers W, Relph K, Harrington K, Melcher A, Roulstone V, Kyula J, Pandha H. Synergistic antitumour effects of rapamycin and oncolytic reovirus. Cancer Gene Ther 2018; 25:148-160. [PMID: 29720674 DOI: 10.1038/s41417-018-0011-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 12/24/2022]
Abstract
There are currently numerous oncolytic viruses undergoing clinical trial evaluation in cancer patients and one agent, Talimogene laherparepvec, has been approved for the treatment of malignant melanoma. This progress highlights the huge clinical potential of this treatment modality, and the focus is now combining these agents with conventional anticancer treatments or agents that enhance viral replication, and thereby oncolysis, in the tumour microenvironment. We evaluated the combination of reovirus with rapamycin in B16F10 cell, a murine model of malignant melanoma, based on potential mechanisms by which mTOR inhibitors might enhance viral oncolysis. Rapamycin was not immunomodulatory in that it had no effect on the generation of an antireovirus-neutralising antibody response in C57/black 6 mice. The cell cycle effects of reovirus (increase G0/G1 fraction) were unaffected by concomitant or sequential exposure of rapamycin. However, rapamycin attenuated viral replication if given prior or concomitantly with reovirus and similarly reduced reovirus-induced apoptotic cell death Annexin V/PI and caspase 3/7 activation studies. We found clear evidence of synergistic antitumour effects of the combination both in vitro and in vivo, which was sequence dependent only in the in vitro setting. In conclusion, we have demonstrated synergistic antitumour efficacy of reovirus and rapamycin combination.
Collapse
Affiliation(s)
- Charles Comins
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Leggett Building, Guildford, Surrey, GU2 7WG, UK
| | - Guy Richard Simpson
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Leggett Building, Guildford, Surrey, GU2 7WG, UK
| | - William Rogers
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Leggett Building, Guildford, Surrey, GU2 7WG, UK
| | - Kate Relph
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Leggett Building, Guildford, Surrey, GU2 7WG, UK
| | - Kevin Harrington
- Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Alan Melcher
- Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Victoria Roulstone
- Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Joan Kyula
- Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Hardev Pandha
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Leggett Building, Guildford, Surrey, GU2 7WG, UK.
| |
Collapse
|
7
|
Bressy C, Majhen D, Raddi N, Jdey W, Cornilleau G, Zig L, Guirouilh-Barbat J, Lopez BS, Bawa O, Opolon P, Grellier E, Benihoud K. Combined therapy of colon carcinomas with an oncolytic adenovirus and valproic acid. Oncotarget 2017; 8:97344-97360. [PMID: 29228615 PMCID: PMC5722567 DOI: 10.18632/oncotarget.22107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 08/04/2017] [Indexed: 12/27/2022] Open
Abstract
The anti-tumor potential of oncolytic adenoviruses (CRAds) has been demonstrated in preclinical and clinical studies. While these agents failed to eradicate tumors when used as a monotherapy, they may be more effective if combined with conventional treatments such as radiotherapy or chemotherapy. This study seeks to evaluate the combination of a CRAd bearing a ∆24 deletion in E1A with valproic acid (VPA), a histone deacetylase inhibitor, for the treatment of human colon carcinomas. This combination led to a strong inhibition of cell growth both in vitro and in vivo compared to treatment with CRAd or VPA alone. This effect did not stem from a better CRAd replication and production in the presence of VPA. Inhibition of cell proliferation and cell death were induced by the combined treatment. Moreover, whereas cells treated only with CRAd displayed a polyploidy (> 4N population), this phenotype was increased in cells treated with both CRAd and VPA. In addition, the increase in polyploidy triggered by combined treatment with CRAd and VPA was associated with the enhancement of H2AX phosphorylation (γH2AX), a hallmark of DNA damage, but also with a decrease of several DNA repair proteins. Finally, viral replication (or E1A expression) was shown to play a key role in the observed effects since no enhancement of polyploidy nor increase in γH2AX were found following cell treatment with a replication-deficient Ad and VPA. Taken together, our results suggest that CRAd and VPA could be used in combination for the treatment of colon carcinomas.
Collapse
Affiliation(s)
- Christian Bressy
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Dragomira Majhen
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Najat Raddi
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Wael Jdey
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Gaétan Cornilleau
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Léna Zig
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Josée Guirouilh-Barbat
- Laboratoire Recombinaison-Réparation et Cancer, UMR 8200 CNRS Stabilité Génétique et Oncogenèse, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Bernard S Lopez
- Laboratoire Recombinaison-Réparation et Cancer, UMR 8200 CNRS Stabilité Génétique et Oncogenèse, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Olivia Bawa
- Unité de pathologie expérimentale de l'IRCIV, Gustave Roussy, Villejuif 94805, France
| | - Paule Opolon
- Unité de pathologie expérimentale de l'IRCIV, Gustave Roussy, Villejuif 94805, France
| | - Elodie Grellier
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Karim Benihoud
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| |
Collapse
|
8
|
Simpson GR, Relph K, Harrington K, Melcher A, Pandha H. Cancer immunotherapy via combining oncolytic virotherapy with chemotherapy: recent advances. Oncolytic Virother 2016; 5:1-13. [PMID: 27579292 PMCID: PMC4996257 DOI: 10.2147/ov.s66083] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oncolytic viruses are multifunctional anticancer agents with huge clinical potential, and have recently passed the randomized Phase III clinical trial hurdle. Both wild-type and engineered viruses have been selected for targeting of specific cancers, to elicit cytotoxicity, and also to generate antitumor immunity. Single-agent oncolytic virotherapy treatments have resulted in modest effects in the clinic. There is increasing interest in their combination with cytotoxic agents, radiotherapy and immune-checkpoint inhibitors. Similarly to oncolytic viruses, the benefits of chemotherapeutic agents may be that they induce systemic antitumor immunity through the induction of immunogenic cell death of cancer cells. Combining these two treatment modalities has to date resulted in significant potential in vitro and in vivo synergies through various mechanisms without any apparent additional toxicities. Chemotherapy has been and will continue to be integral to the management of advanced cancers. This review therefore focuses on the potential for a number of common cytotoxic agents to be combined with clinically relevant oncolytic viruses. In many cases, this combined approach has already advanced to the clinical trial arena.
Collapse
Affiliation(s)
- Guy R Simpson
- Department of Clinical and Experimental Medicine, Targeted Cancer Therapy, Faculty of Health and Medical Sciences, University of Surrey, Guildford
| | - Kate Relph
- Department of Clinical and Experimental Medicine, Targeted Cancer Therapy, Faculty of Health and Medical Sciences, University of Surrey, Guildford
| | - Kevin Harrington
- Targeted Therapy, The Institute of Cancer Research/The Royal Marsden NIHR Biomedical Research Centre, London
| | - Alan Melcher
- Targeted and Biological Therapies, Oncology and Clinical Research, Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Hardev Pandha
- Department of Clinical and Experimental Medicine, Targeted Cancer Therapy, Faculty of Health and Medical Sciences, University of Surrey, Guildford
| |
Collapse
|
9
|
Nande R, Howard CM, Claudio PP. Ultrasound-mediated oncolytic virus delivery and uptake for increased therapeutic efficacy: state of art. Oncolytic Virother 2015; 4:193-205. [PMID: 27512682 PMCID: PMC4918399 DOI: 10.2147/ov.s66097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The field of ultrasound (US) has changed significantly from medical imaging and diagnosis to treatment strategies. US contrast agents or microbubbles (MB) are currently being used as potential carriers for chemodrugs, small molecules, nucleic acids, small interfering ribonucleic acid, proteins, adenoviruses, and oncolytic viruses. Oncolytic viruses can selectively replicate within and destroy a cancer cell, thus making them a powerful therapeutic in treating late-stage or metastatic cancer. These viruses have been shown to have robust activity in clinical trials when injected directly into tumor nodules. However limitations in oncolytic virus’ effectiveness and its delivery approach have warranted exploration of ultrasound-mediated delivery. Gene therapy bearing adenoviruses or oncolytic viruses can be coupled with MBs and injected intravenously. Following application of US energy to the target region, the MBs cavitate, and the resulting shock wave enhances drug, gene, or adenovirus uptake. Though the underlying mechanism is yet to be fully understood, there is evidence to suggest that mechanical pore formation of cellular membranes allows for the temporary uptake of drugs. This delivery method circumvents the limitations due to stimulation of the immune system that prevented intravenous administration of viruses. This review provides insight into this intriguing new frontier on the delivery of oncolytic viruses to tumor sites.
Collapse
Affiliation(s)
- Rounak Nande
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - Candace M Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pier Paolo Claudio
- Department of BioMolecular Sciences and National Center for Natural Products Research, School of Pharmacy, University of Mississippi, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
10
|
Nguyen A, Ho L, Wan Y. Chemotherapy and Oncolytic Virotherapy: Advanced Tactics in the War against Cancer. Front Oncol 2014; 4:145. [PMID: 24967214 PMCID: PMC4052116 DOI: 10.3389/fonc.2014.00145] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/28/2014] [Indexed: 01/10/2023] Open
Abstract
Cancer is a traitorous archenemy that threatens our survival. Its ability to evade detection and adapt to various cancer therapies means that it is a moving target that becomes increasingly difficult to attack. Through technological advancements, we have developed sophisticated weapons to fight off tumor growth and invasion. However, if we are to stand a chance in this war against cancer, advanced tactics will be required to maximize the use of our available resources. Oncolytic viruses (OVs) are multi-functional cancer-fighters that can be engineered to suit many different strategies; in particular, their retooling can facilitate increased capacity for direct tumor killing (oncolytic virotherapy) and elicit adaptive antitumor immune responses (oncolytic immunotherapy). However, administration of these modified OVs alone, rarely induces successful regression of established tumors. This may be attributed to host antiviral immunity that acts to eliminate viral particles, as well as the capacity for tumors to adapt to therapeutic selective pressure. It has been shown that various chemotherapeutic drugs with distinct functional properties can potentiate the antitumor efficacy of OVs. In this review, we summarize the chemotherapeutic combinatorial strategies used to optimize virally induced destruction of tumors. With a particular focus on pharmaceutical immunomodulators, we discuss how specific therapeutic contexts may alter the effects of these synergistic combinations and their implications for future clinical use.
Collapse
Affiliation(s)
- Andrew Nguyen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, ON , Canada
| | - Louisa Ho
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, ON , Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, ON , Canada
| |
Collapse
|
11
|
Bressy C, Benihoud K. Association of oncolytic adenoviruses with chemotherapies: an overview and future directions. Biochem Pharmacol 2014; 90:97-106. [PMID: 24832861 DOI: 10.1016/j.bcp.2014.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/03/2014] [Accepted: 05/05/2014] [Indexed: 12/12/2022]
Abstract
Oncolytic adenoviruses have been used in different preclinical and clinical studies, showing their capacity to kill tumor cells without major adverse events. However, these studies also underline the limitations of this approach. The efficacy of oncolytic adenoviruses is hampered by their limited ability to transduce some tumor types, their lack of selectivity, and their poor dissemination within tumors. In addition, the host immune response may limit oncolytic adenovirus efficacy. Combining oncolytic adenoviruses with chemotherapeutics constitutes an appealing strategy to increase their potency. The first part of this review describes the molecular basis of oncolytic adenoviruses, their use in preclinical studies and clinical trials, their limitations, and strategies to circumvent these limitations. The second part will focus on studies combining oncolytic adenoviruses with chemotherapeutic drugs, including standard chemotherapeutic drugs, molecularly targeted drugs, and other drugs that have been combined with oncolytic adenoviruses. Finally, based on these studies, we describe future directions and general rules that could be followed to identify chemotherapeutic drugs displaying additive/synergistic effects when combined with oncolytic adenoviruses.
Collapse
Affiliation(s)
- Christian Bressy
- CNRS UMR 8203, Vectorologie et thérapeutiques anti-cancéreuses, Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif Cedex, France; Univ Paris-Sud, 15 rue Georges Clémenceau, 91405 Orsay Cedex, France
| | - Karim Benihoud
- CNRS UMR 8203, Vectorologie et thérapeutiques anti-cancéreuses, Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif Cedex, France; Univ Paris-Sud, 15 rue Georges Clémenceau, 91405 Orsay Cedex, France.
| |
Collapse
|
12
|
Ilkow CS, Swift SL, Bell JC, Diallo JS. From scourge to cure: tumour-selective viral pathogenesis as a new strategy against cancer. PLoS Pathog 2014; 10:e1003836. [PMID: 24453963 PMCID: PMC3894191 DOI: 10.1371/journal.ppat.1003836] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Tumour mutations corrupt cellular pathways, and accumulate to disrupt, dysregulate, and ultimately avoid mechanisms of cellular control. Yet the very changes that tumour cells undergo to secure their own growth success also render them susceptible to viral infection. Enhanced availability of surface receptors, disruption of antiviral sensing, elevated metabolic activity, disengagement of cell cycle controls, hyperactivation of mitogenic pathways, and apoptotic avoidance all render the malignant cell environment highly supportive to viral replication. The therapeutic use of oncolytic viruses (OVs) with a natural tropism for infecting and subsequently lysing tumour cells is a rapidly progressing area of cancer research. While many OVs exhibit an inherent degree of tropism for transformed cells, this can be further promoted through pharmacological interventions and/or the introduction of viral mutations that generate recombinant oncolytic viruses adapted to successfully replicate only in a malignant cellular environment. Such adaptations that augment OV tumour selectivity are already improving the therapeutic outlook for cancer, and there remains tremendous untapped potential for further innovation.
Collapse
Affiliation(s)
- Carolina S. Ilkow
- Centre for Innovative Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, Ontario, Canada
| | | | - John C. Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
13
|
Li X, Tong LJ, Ding J, Meng LH. Systematic combination screening reveals synergism between rapamycin and sunitinib against human lung cancer. Cancer Lett 2014; 342:159-66. [DOI: 10.1016/j.canlet.2013.08.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/29/2013] [Accepted: 08/31/2013] [Indexed: 10/26/2022]
|
14
|
Jiang ZK, Johnson M, Moughon DL, Kuo J, Sato M, Wu L. Rapamycin enhances adenovirus-mediated cancer imaging and therapy in pre-immunized murine hosts. PLoS One 2013; 8:e73650. [PMID: 24023896 PMCID: PMC3759448 DOI: 10.1371/journal.pone.0073650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/19/2013] [Indexed: 12/24/2022] Open
Abstract
Tumor-specific adenoviral vectors comprise a fruitful gene-based diagnostic imaging and therapy research area for advanced stage of cancer, including metastatic disease. However, clinical translation of viral vectors has encountered considerable obstacles, largely due to host immune responses against the virus. Here, we explored the utilization of an immunosuppressant, rapamycin, to circumvent the anti-adenovirus immunity in immunocompetent murine prostate cancer models. Rapamycin diminished adenoviral-induced acute immune response by inhibiting NF-κB activation; it also reduced the scale and delayed the onset of inflammatory cytokine secretion. Further, we found that rapamycin abrogated anti-adenovirus antibody production and retarded the function of myeloid cells and lymphocytes that were activated upon viral administration in pre-immunized hosts. Thus, the co-administration of rapamycin prolonged and enhanced adenovirus-delivered transgene expression in vivo, and thereby augmented the imaging capability of adenoviral vectors in both bioluminescent and positron emission tomography modalities. Furthermore, we showed that despite an excellent response of cancer cells to a cytotoxic gene therapeutic vector in vitro, only minimal therapeutic effects were observed in vivo in pre-immunized mice. However, when we combined gene therapy with transient immunosuppression, complete tumor growth arrest was achieved. Overall, transient immunosuppression by rapamycin was able to boost the diagnostic utility and therapeutic potentials of adenoviral vectors.
Collapse
Affiliation(s)
- Ziyue Karen Jiang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Institute of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Mai Johnson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Institute of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Diana L. Moughon
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Institute of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Jennifer Kuo
- Department of Molecular, Cellular and Developmental Biology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Makoto Sato
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Institute of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Institute of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
Bazan-Peregrino M, Rifai B, Carlisle RC, Choi J, Arvanitis CD, Seymour LW, Coussios CC. Cavitation-enhanced delivery of a replicating oncolytic adenovirus to tumors using focused ultrasound. J Control Release 2013; 169:40-7. [PMID: 23562636 DOI: 10.1016/j.jconrel.2013.03.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/18/2013] [Indexed: 10/27/2022]
Abstract
Oncolytic viruses (OV) and ultrasound-enhanced drug delivery are powerful novel technologies. OV selectively self-amplify and kill cancer cells but their clinical use has been restricted by limited delivery from the bloodstream into the tumor. Ultrasound has been previously exploited for targeted release of OV in vivo, but its use to induce cavitation, microbubble oscillations, for enhanced OV tumor extravasation and delivery has not been previously reported. By identifying and optimizing the underlying physical mechanism, this work demonstrates that focused ultrasound significantly enhances the delivery and biodistribution of systemically administered OV co-injected with microbubbles. Up to a fiftyfold increase in tumor transgene expression was achieved, without any observable tissue damage. Ultrasound exposure parameters were optimized as a function of tumor reperfusion time to sustain inertial cavitation, a type of microbubble activity, throughout the exposure. Passive detection of acoustic emissions during treatment confirmed inertial cavitation as the mechanism responsible for enhanced delivery and enabled real-time monitoring of successful viral delivery.
Collapse
Affiliation(s)
- Miriam Bazan-Peregrino
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX3 7DQ, UK.
| | | | | | | | | | | | | |
Collapse
|
16
|
Angarita FA, Acuna SA, Ottolino-Perry K, Zerhouni S, McCart JA. Mounting a strategic offense: fighting tumor vasculature with oncolytic viruses. Trends Mol Med 2013; 19:378-92. [PMID: 23540715 DOI: 10.1016/j.molmed.2013.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/23/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
Blood supply within a tumor drives progression and ultimately allows for metastasis. Many anticancer therapies target tumor vasculature, but their individual effectiveness is limited because they induce indirect cell death. Agents that disrupt nascent and/or established tumor vasculature while simultaneously killing cancer cells would certainly have a greater impact. Oncolytic virotherapy utilizes attenuated viruses that replicate specifically within a tumor. They induce cytotoxicity through a combination of direct cell lysis, antitumor immune stimulation, and recently identified antitumor vascular effects. This review summarizes the novel preclinical and clinical evidence regarding the antitumor vascular effects of oncolytic viruses, which include infection and lysis of tumor endothelial cells, natural or genetically engineered antiangiogenic properties, and combination therapy with clinically approved antivascular agents.
Collapse
Affiliation(s)
- Fernando A Angarita
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2M1 Canada
| | | | | | | | | |
Collapse
|
17
|
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
|
18
|
Meng S, Xu J, Wu Y, Ding C. Targeting autophagy to enhance oncolytic virus-based cancer therapy. Expert Opin Biol Ther 2013; 13:863-73. [PMID: 23488666 DOI: 10.1517/14712598.2013.774365] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Autophagy is a conserved catabolic process crucial in maintaining cellular homeostasis. On infection, oncolytic viruses (OVs) perturb the cellular autophagy machinery in infected tumor cells both in vitro and in vivo. Currently, pharmacological modulation of autophagy in OV-infected tumor cells has been shown to augment OV-mediated antitumor effects in preclinical studies. Combination of OVs with autophagy modulators can, therefore, have many potential applications in the future research on targeting autophagy and novel anticancer therapies. AREAS COVERED This review provides a detailed description of known interactions between OVs and autophagy and summarizes the roles of autophagy in OV replication and cell lysis. The recent literature on targeting autophagy with either the autophagy inducers, such as rapamycin, or autophagy inhibitors, such as chloroquine, to increase OV-induced cytotoxicity is reviewed to help researchers in further investigations. The major challenge for investigators is to understand the molecular mechanism underlying the interplay between OV and the autophagy machinery and its effect on oncolysis. EXPERT OPINION Targeting the cellular autophagy machinery could be explored as a new therapeutic strategy to enhance OV-mediated antitumor effects in the future.
Collapse
Affiliation(s)
- Songshu Meng
- Dalian Medical University Cancer Center, Institute of Cancer Stem Cell, 9 Lvshun Road South, Dalian 116044, Chin.
| | | | | | | |
Collapse
|
19
|
Jiang ZK, Koh SBS, Sato M, Atanasov IC, Johnson M, Zhou ZH, Deming TJ, Wu L. Engineering polypeptide coatings to augment gene transduction and in vivo stability of adenoviruses. J Control Release 2013; 166:75-85. [PMID: 23247040 DOI: 10.1016/j.jconrel.2012.10.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/19/2012] [Accepted: 10/29/2012] [Indexed: 01/19/2023]
Abstract
We sought to modify adenoviral (Ad) particles by incorporating the advantageous characteristics of non-viral gene delivery vehicles to complement the viral vectors. α-Amino acid-N-carboxyanhydride chemistry was used to synthesize homopolypeptides and diblock copolypeptides that possess well-defined secondary structures. Using cryo-electron and fluorescence microscopy, we showed that these polypeptides can coat the surfaces of Ad particles in a non-covalent manner to modify their transduction properties. The coated Ad particles were found to bind to and be internalized by cells. In contrast to reports using covalently PEGylated Ad particles, we found that our physically coated Ad hybrid complexes facilitate gene transfer both in vitro and in vivo. We showed that our polypeptide coating was able to shield the Ad particles from the neutralizing effect of antibodies and mitigate the binding of blood coagulation factor (Factor X) in vitro. The coating also reduced the antigenicity of Ad in immunocompetent mice. The biodistribution of the systemically administered hybrid complexes mirrored the behavior of both viral and non-viral vectors, exhibiting liver tropism as well as enhanced lung transduction. These data demonstrated that our non-covalent modification was able to alter Ad's interactions with cells and organs with retention of transduction efficiency. Advantages such as facile coating of the Ad vector, design flexibility and ease of attaching ligands to the polypeptides make this system potentially useful as a platform for adding functionalities to Ad to target cancer metastasis.
Collapse
Affiliation(s)
- Ziyue Karen Jiang
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Jiang G, Zhang L, Xin Y, Pei DS, Wei ZP, Liu YQ, Zheng JN. Conditionally replicating adenoviruses carrying mda-7/IL-24 for cancer therapy. Acta Oncol 2012; 51:285-92. [PMID: 21995527 DOI: 10.3109/0284186x.2011.621447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) suppresses growth and induces apoptosis in a broad range of human cancers without significant cytotoxicity to normal cells. Conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. METHODS This review provides an overview of specifications for a novel anti-tumor approach CRAds carrying IL-24, and discusses recent progress in this field. RESULTS Studies in multiple laboratories report that CRAds carrying IL-24 selectively induced apoptosis in some cancer cells, and enhanced selective toxicity to cancer cells when combined with chemotherapeutic agents. CONCLUSION CRAds carrying IL-24 may prove a novel and effective approach for the treatment of cancers.
Collapse
Affiliation(s)
- Guan Jiang
- Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Gomez-Manzano C, Jiang H, Alonso M, Yung WKA, Fueyo J. Gene therapy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:331-8. [PMID: 22230451 DOI: 10.1016/b978-0-444-52138-5.00021-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Candelaria Gomez-Manzano
- Department of Neuro-oncology, The University of Texas, M. D Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | |
Collapse
|
22
|
GALV expression enhances the therapeutic efficacy of an oncolytic adenovirus by inducing cell fusion and enhancing virus distribution. Gene Ther 2011; 19:1048-57. [PMID: 22113313 DOI: 10.1038/gt.2011.184] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The limitations of the current oncolytic adenoviruses for cancer therapy include insufficient potency and poor distribution of the virus throughout the tumor mass. To address these problems, we generated an oncolytic adenovirus expressing the hyperfusogenic form of the gibbon-ape leukemia virus (GALV) envelope glycoprotein under the control of the adenovirus major late promoter. The oncolytic properties of the new fusogenic adenovirus, ICOVIR16, were analyzed both in vitro and in vivo, and compared with that of its non-fusogenic counterpart, ICOVIR15. Our results indicate that GALV expression by ICOVIR16 induced extensive syncytia formation and enhanced tumor cell killing in a variety of tumor cell types. When injected intratumorally or intravenously into mice with large pre-established melanoma or pancreatic tumors, ICOVIR16 rapidly reduced tumor burden, and in some cases, resulted in complete eradication of the tumors. Importantly, GALV expression induced tumor cell fusion in vivo and enhanced the spreading of the virus throughout the tumor. Taken together, these results indicate that GALV expression can improve the antitumoral potency of an oncolytic adenovirus and suggest that ICOVIR16 is a promising candidate for clinical evaluation in patients with cancer.
Collapse
|
23
|
Reovirus therapy stimulates endoplasmic reticular stress, NOXA induction, and augments bortezomib-mediated apoptosis in multiple myeloma. Oncogene 2011; 31:3023-38. [PMID: 22002308 DOI: 10.1038/onc.2011.478] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Oncolytic virotherapy with reovirus has demonstrated anti-cancer activity and minimal toxicity in clinical trials, but the mechanisms underlying these effects have not been fully elucidated. Reolysin, a proprietary formulation of reovirus for cancer therapy, stimulated selective viral replication and apoptosis in multiple myeloma (MM) cells. Reolysin-mediated apoptosis was associated with an induction of endoplasmic reticular (ER) stress-related gene expression, swelling of the endoplasmic reticulum, increases in intracellular calcium levels and a strong induction of the Bcl-2 homology 3 (BH3)-only pro-apoptotic protein NOXA. Knockdown of NOXA expression by short hairpin RNA significantly reduced the pro-apoptotic effects of Reolysin. We next showed that co-administration of Reolysin and bortezomib resulted in the dual accumulation of viral and ubiquitinated proteins, which led to enhanced ER stress, NOXA induction and apoptosis. Importantly, the combination of reovirus infection and proteasomal inhibition significantly decreased tumor burden in a xenograft and syngeneic bone disease model of MM without exhibiting adverse side effects. Our study establishes ER stress stimulation and NOXA induction as novel mediators of reovirus-induced apoptosis. Furthermore, reovirus infection can be used as a promising approach to augment the anti-myeloma activity of bortezomib by promoting additional stress to the endoplasmic reticulum of MM cells.
Collapse
|
24
|
Bazan-Peregrino M, Arvanitis CD, Rifai B, Seymour LW, Coussios CC. Ultrasound-induced cavitation enhances the delivery and therapeutic efficacy of an oncolytic virus in an in vitro model. J Control Release 2011; 157:235-42. [PMID: 21982902 DOI: 10.1016/j.jconrel.2011.09.086] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 09/21/2011] [Accepted: 09/23/2011] [Indexed: 01/06/2023]
Abstract
We investigated whether ultrasound-induced cavitation at 0.5 MHz could improve the extravasation and distribution of a potent breast cancer-selective oncolytic adenovirus, AdEHE2F-Luc, to tumour regions that are remote from blood vessels. We developed a novel tumour-mimicking model consisting of a gel matrix containing human breast cancer cells traversed by a fluid channel simulating a tumour blood vessel, through which the virus and microbubbles could be made to flow. Ultrasonic pressures were chosen to maximize either broadband emissions, associated with inertial cavitation, or ultraharmonic emissions, associated with stable cavitation, while varying duty cycle to keep the total acoustic energy delivered constant for comparison across exposures. None of the exposure conditions tested affected cell viability in the absence of the adenovirus. When AdEHE2F-Luc was delivered via the vessel, inertial cavitation increased transgene expression in tumour cells by up to 200 times. This increase was not observed in the absence of Coxsackie and Adenovirus Receptor cell expression, discounting sonoporation as the mechanism of action. In the presence of inertial cavitation, AdEHE2F-Luc distribution was greatly improved in the matrix surrounding the vessel, particularly in the direction of the ultrasound beam; this enabled AdEHE2F-Luc to kill up to 80% of cancer cells within the ultrasound focal volume in the gel 24 hours after delivery, compared to 0% in the absence of cavitation.
Collapse
Affiliation(s)
- Miriam Bazan-Peregrino
- Institute of Biomedical Engineering, Department of Engineering Science, Old Road Campus Research Building, University of Oxford, Headington, Oxford OX3 7DQ, United Kingdom.
| | | | | | | | | |
Collapse
|
25
|
Jiang ZK, Sato M, Wei LH, Kao C, Wu L. Androgen-independent molecular imaging vectors to detect castration-resistant and metastatic prostate cancer. Cancer Res 2011; 71:6250-60. [PMID: 21933883 DOI: 10.1158/0008-5472.can-11-1520] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prostate-specific promoters are frequently employed in gene-mediated molecular imaging and therapeutic vectors to diagnose and treat castration-resistant prostate cancer (CRPC) that emerges from hormone ablation therapy. Many of the conventional prostate-specific promoters rely on the androgen axis to drive gene expression. However, considering the cancer heterogeneity and varying androgen receptor status, we herein evaluated the utility of prostate-specific enhancing sequence (PSES), an androgen-independent promoter in CRPC. The PSES is a fused enhancer derived from the prostate-specific antigen (PSA) and prostate-specific membrane antigen gene regulatory region. We augmented the activity of PSES by the two-step transcriptional amplification (TSTA) system to drive the expression of imaging reporter genes for either bioluminescent or positron emission tomography (PET) imaging. The engineered PSES-TSTA system exhibits greatly elevated transcriptional activity, androgen independency, and strong prostate specificity, verified in cell culture and preclinical animal experimentations. These advantageous features of PSES-TSTA elicit superior gene expression capability for CRPC in comparison with the androgen-dependent PSA promoter-driven system. In preclinical settings, we showed robust PET imaging capacity of PSES-TSTA in a castrated prostate xenograft model. Moreover, intravenous administrated PSES-TSTA bioluminescent vector correctly identified tibial bone marrow metastases in 9 of 9 animals, whereas NaF- and FDG-PET was unable to detect the lesions. Taken together, this study showed the promising utility of a potent, androgen-independent, and prostate cancer-specific expression system in directing gene-based molecular imaging in CRPC, even in the context of androgen deprivation therapy.
Collapse
Affiliation(s)
- Ziyue Karen Jiang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
26
|
Thomas DL, Doty R, Tosic V, Liu J, Kranz DM, McFadden G, Macneill AL, Roy EJ. Myxoma virus combined with rapamycin treatment enhances adoptive T cell therapy for murine melanoma brain tumors. Cancer Immunol Immunother 2011; 60:1461-72. [PMID: 21656158 DOI: 10.1007/s00262-011-1045-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 05/20/2011] [Indexed: 12/11/2022]
Abstract
Adoptive transfer of tumor-specific T cells has shown some success for treating metastatic melanoma. We evaluated a novel strategy to improve adoptive therapy by administering both T cells and oncolytic myxoma virus to mice with syngeneic B16.SIY melanoma brain tumors. Adoptive transfer of activated CD8(+) 2C T cells that recognize SIY peptide doubled survival time, but SIY-negative tumors recurred. Myxoma virus killed B16.SIY cells in vitro, and intratumoral injection of virus led to selective and transient infection of the tumor. Virus treatment recruited innate immune cells to the tumor and induced IFNβ production in the brain, resulting in limited oncolytic effects in vivo. To counter this, we evaluated the safety and efficacy of co-administering 2C T cells, myxoma virus, and either rapamycin or neutralizing antibodies against IFNβ. Mice that received either triple combination therapy survived significantly longer with no apparent side effects, but eventually relapsed. Importantly, rapamycin treatment did not impair T cell-mediated tumor destruction, supporting the feasibility of combining adoptive immunotherapy and rapamycin-enhanced virotherapy. Myxoma virus may be a useful vector for transient delivery of therapeutic genes to a tumor to enhance T cell responses.
Collapse
Affiliation(s)
- Diana L Thomas
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Jiang G, Xin Y, Zheng JN, Liu YQ. Combining conditionally replicating adenovirus-mediated gene therapy with chemotherapy: a novel antitumor approach. Int J Cancer 2011; 129:263-74. [PMID: 21509783 DOI: 10.1002/ijc.25948] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 01/05/2011] [Indexed: 12/16/2022]
Abstract
Despite significant improvements in diagnosis and innovations in the therapy of specific cancers, effective treatment of neoplastic diseases still presents major challenges. Recent studies have shown that conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. Several studies in animal models have demonstrated that the combination of CRAds-mediated gene therapy and chemotherapy has greater therapeutic benefit than either treatment modality alone. In this review, an overview of specifications for a novel antitumor approach combining CRAd-gene therapy and chemotherapy is provided and recent progress in this field is discussed.
Collapse
Affiliation(s)
- Guan Jiang
- Center for Disease Control and Prevention of Xuzhou City, Xuzhou 221006, China
| | | | | | | |
Collapse
|
28
|
Lin CI, Whang EE, Donner DB, Du J, Lorch J, He F, Jiang X, Price BD, Moore FD, Ruan DT. Autophagy induction with RAD001 enhances chemosensitivity and radiosensitivity through Met inhibition in papillary thyroid cancer. Mol Cancer Res 2010; 8:1217-26. [PMID: 20736296 DOI: 10.1158/1541-7786.mcr-10-0162] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although autophagy is generally considered a prosurvival mechanism that preserves viability, there is evidence that it could drive an alternative programmed cell death pathway in cells with defects in apoptosis. Because the inhibition of autophagic activity promotes resistance to both chemotherapy and external beam radiation in papillary thyroid cancer (PTC), we determined if RAD001, a potent activator of autophagy, improves the efficacy of either therapy. We found that RAD001 increased the expression level of light chain 3-II, a marker for autophagy, as well as autophagosome formation in cell lines and in human PTC ex vivo. RAD001 sensitized PTC to doxorubicin and external beam radiation in a synergistic fashion, suggesting that combination therapy could improve therapeutic response at less toxic concentrations. The effects of RAD001 were abrogated by RNAi knockdown of the autophagy-related gene 5, suggesting that RAD001 acts, in part, by enhancing autophagy. Because the synergistic activity of RAD001 with doxorubicin and external radiation suggests distinct and complementary mechanisms of action, we characterized how autophagy modulates signaling pathways in PTC. To do so, we performed kinome profiling and discovered that autophagic activation resulted in Src phosphorylation and Met dephosphorylation. Src inhibition did not reverse the effects of RAD001, whereas Met inhibition reversed the effects of autophagy blockade on chemosensitivity. These results suggest that the anticancer effects of autophagic activation are mediated largely through Met. We conclude that RAD001 induces autophagy, which enhances the therapeutic response to cytotoxic chemotherapy and external beam radiation in PTC.
Collapse
Affiliation(s)
- Chi-Iou Lin
- Department of Surgery, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Taguchi F, Kodera Y, Katanasaka Y, Yanagihara K, Tamura T, Koizumi F. Efficacy of RAD001 (everolimus) against advanced gastric cancer with peritoneal dissemination. Invest New Drugs 2010; 29:1198-205. [DOI: 10.1007/s10637-010-9464-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 05/20/2010] [Indexed: 12/25/2022]
|
30
|
Vesicular stomatitis virus oncolysis is potentiated by impairing mTORC1-dependent type I IFN production. Proc Natl Acad Sci U S A 2010; 107:1576-81. [PMID: 20080710 DOI: 10.1073/pnas.0912344107] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Oncolytic viruses constitute a promising therapy against malignant gliomas (MGs). However, virus-induced type I IFN greatly limits its clinical application. The kinase mammalian target of rapamycin (mTOR) stimulates type I IFN production via phosphorylation of its effector proteins, 4E-BPs and S6Ks. Here we show that mouse embryonic fibroblasts and mice lacking S6K1 and S6K2 are more susceptible to vesicular stomatitis virus (VSV) infection than their WT counterparts as a result of an impaired type I IFN response. We used this knowledge to employ a pharmacoviral approach to treat MGs. The highly specific inhibitor of mTOR rapamycin, in combination with an IFN-sensitive VSV-mutant strain (VSV(DeltaM51)), dramatically increased the survival of immunocompetent rats bearing MGs. More importantly, VSV(DeltaM51) selectively killed tumor, but not normal cells, in MG-bearing rats treated with rapamycin. These results demonstrate that reducing type I IFNs through inhibition of mTORC1 is an effective strategy to augment the therapeutic activity of VSV(DeltaM51).
Collapse
|
31
|
Peerlinck I, Merron A, Baril P, Conchon S, Martin-Duque P, Hindorf C, Burnet J, Quintanilla M, Hingorani M, Iggo R, Lemoine NR, Harrington K, Vassaux G. Targeted radionuclide therapy using a Wnt-targeted replicating adenovirus encoding the Na/I symporter. Clin Cancer Res 2009; 15:6595-601. [PMID: 19861465 DOI: 10.1158/1078-0432.ccr-09-0262] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE The Na/I symporter (hNIS) promotes concentration of iodine in cells. In cancer gene therapy, this transgene has potential as a reporter gene for molecular imaging of viral biodistribution and as a therapeutic protein promoting (131)I-mediated radiotherapy. Here, we combined the imaging and therapeutic potential of hNIS in an oncolytic adenoviruses targeting colorectal cancer cells. EXPERIMENTAL DESIGN We generated an adenovirus (AdIP2) encoding hNIS and capable of selective replication in colorectal carcinoma cells. The selectivity of this virus was verified in vitro and in vivo. Its spread in tumors was monitored in vivo using single-photon emission computed tomography/CT imaging upon (99m)TcO(4)(-) injection and confirmed by immunohistochemistry. Metabolic radiotherapy was done through injection of therapeutic doses of (131)I(-). RESULTS We showed in vitro and in vivo the selectivity of AdIP2 and that hNIS expression is restricted to the target cells. Imaging and immunohistochemical data showed that viral spread is limited and that the point of maximal hNIS expression is reached 48 hours after a single intratumoral injection. Administration of a single therapeutic dose of (131)I at this time point led to a dramatic reduction in tumor size not observed in hNIS-negative viruses. CONCLUSIONS This report showed for the first time that the combination of the imaging and therapeutic potentials of hNIS can be applied to oncolytic adenoviruses in experimental models of cancer.
Collapse
Affiliation(s)
- Inge Peerlinck
- Centre for Molecular Oncology, Institute of Cancer, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Baril P, Martin-Duque P, Vassaux G. Visualization of gene expression in the live subject using the Na/I symporter as a reporter gene: applications in biotherapy. Br J Pharmacol 2009; 159:761-71. [PMID: 19814733 DOI: 10.1111/j.1476-5381.2009.00412.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Biotherapies involve the utilization of antibodies, genetically modified viruses, bacteria or cells for therapeutic purposes. Molecular imaging has the potential to provide unique information that will guarantee their biosafety in humans and provide a rationale for the future development of new generations of reagents. In this context, non-invasive imaging of gene expression is an attractive prospect, allowing precise, spacio-temporal measurements of gene expression in longitudinal studies involving gene transfer vectors. With the emergence of cell therapies in regenerative medicine, it is also possible to track cells injected into subjects. In this context, the Na/I symporter (NIS) has been used in preclinical studies. Associated with a relevant radiotracer ((123)I(-), (124)I(-), (99m)TcO4(-)), NIS can be used to monitor gene transfer and the spread of selectively replicative viruses in tumours as well as in cells with a therapeutic potential. In addition to its imaging potential, NIS can be used as a therapeutic transgene through its ability to concentrate therapeutic doses of radionuclides in target cells. This dual property has applications in cancer treatment and could also be used to eradicate cells with therapeutic potential in the case of adverse events. Through experience acquired in preclinical studies, we can expect that non-invasive molecular imaging using NIS as a transgene will be pivotal for monitoring in vivo the exact distribution and pharmacodynamics of gene expression in a precise and quantitative way. This review highlights the applications of NIS in biotherapy, with a particular emphasis on image-guided radiotherapy, monitoring of gene and vector biodistribution and trafficking of stem cells.
Collapse
Affiliation(s)
- Patrick Baril
- Inserm U948, Université de Nantes, Nantes Atlantique Universités, EA4274, Institut des Maladies de l'Appareil Digestif, CHU Hôtel Dieu, Nantes, France.
| | | | | |
Collapse
|
33
|
Alvarez-Breckenridge C, Kaur B, Chiocca EA. Pharmacologic and chemical adjuvants in tumor virotherapy. Chem Rev 2009; 109:3125-40. [PMID: 19462957 DOI: 10.1021/cr900048k] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Christopher Alvarez-Breckenridge
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center and The Ohio State University Medical Center, Columbus, Ohio, USA
| | | | | |
Collapse
|
34
|
Cafferata EG, Macció DR, Lopez MV, Viale DL, Carbone C, Mazzolini G, Podhajcer OL. A novel A33 promoter-based conditionally replicative adenovirus suppresses tumor growth and eradicates hepatic metastases in human colon cancer models. Clin Cancer Res 2009; 15:3037-49. [PMID: 19336523 DOI: 10.1158/1078-0432.ccr-08-1161] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A33 antigen is a membrane-bound protein expressed in intestinal epithelium that is overexpressed in 95% of primary and metastatic colorectal carcinomas but is absent in most epithelial tissues and tumor types. We hypothesized that A33 promoter might be useful in the design of a conditionally replicative adenovirus for the treatment of colorectal cancer (CRC). EXPERIMENTAL DESIGN We cloned an A33 promoter fragment (A33Pr) that extends from -105 to +307 bp. Using luciferase activity as a reporter gene, we showed that A33Pr was active in CRC cell lines. We next constructed a conditionally replicative adenovirus named AV22EL where E1A was placed under the control of A33Pr. The tumor-specific oncolytic effect of AV22EL was investigated both in vitro and in vivo. RESULTS AV22EL induced specific in vitro lysis of human CRC cell lines that expressed A33 and have negligible lytic capacity on cells that lacked or had minimal A33 expression, including normal human colonic cells. In vivo, a marked reduction of tumor growth and increased long-term survival rates were observed in nude mice xenografted with s.c. CRC tumors. Combination with 5-fluorouracil induced an additive effect in vitro with no toxic effects in vivo. Remarkably, AV22EL completely eliminated established hepatic metastases in >90% of mice and restored hepatic function according to biochemical parameters. Its systemic administration induced E1A expression only in the hepatic metastasis but not in normal organs. CONCLUSIONS These data show that AV22EL is a stringently regulated and potent oncolytic agent for the treatment of CRC.
Collapse
Affiliation(s)
- Eduardo G Cafferata
- Laboratorio de Terapia Molecular y Celular, Instituto Leloir and Instituto de Investigaciones Bioquimicas de Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
35
|
Haseley A, Alvarez-Breckenridge C, Chaudhury AR, Kaur B. Advances in oncolytic virus therapy for glioma. ACTA ACUST UNITED AC 2009; 4:1-13. [PMID: 19149710 DOI: 10.2174/157488909787002573] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The World Health Organization grossly classifies the various types of astrocytomas using a grade system with grade IV gliomas having the worst prognosis. Oncolytic virus therapy is a novel treatment option for GBM patients. Several patents describe various oncolytic viruses used in preclinical and clinical trials to evaluate safety and efficacy. These viruses are natural or genetically engineered from different viruses such as HSV-1, Adenovirus, Reovirus, and New Castle Disease Virus. While several anecdotal studies have indicated therapeutic advantage, recent clinical trials have revealed the safety of their usage, but demonstration of significant efficacy remains to be established. Oncolytic viruses are being redesigned with an interest in combating the tumor microenvironment in addition to defeating the cancerous cells. Several patents describe the inclusion of tumor microenvironment modulating genes within the viral backbone and in particular those which attack the tumor angiotome. The very innovative approaches being used to improve therapeutic efficacy include: design of viruses which can express cytokines to activate a systemic antitumor immune response, inclusion of angiostatic genes to combat tumor vasculature, and also enzymes capable of digesting tumor extra cellular matrix (ECM) to enhance viral spread through solid tumors. As increasingly more novel viruses are being tested and patented, the future battle against glioma looks promising.
Collapse
Affiliation(s)
- Amy Haseley
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, College of Medicine, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
36
|
Carlisle RC, Di Y, Cerny AM, Sonnen AFP, Sim RB, Green NK, Subr V, Ulbrich K, Gilbert RJC, Fisher KD, Finberg RW, Seymour LW. Human erythrocytes bind and inactivate type 5 adenovirus by presenting Coxsackie virus-adenovirus receptor and complement receptor 1. Blood 2009; 113:1909-18. [PMID: 19131551 PMCID: PMC2651010 DOI: 10.1182/blood-2008-09-178459] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 11/07/2008] [Indexed: 12/31/2022] Open
Abstract
Type 5 adenovirus (Ad5) is a human pathogen that has been widely developed for therapeutic uses, with only limited success to date. We report here the novel finding that human erythrocytes present Coxsackie virus-adenovirus receptor (CAR) providing an Ad5 sequestration mechanism that protects against systemic infection. Interestingly, erythrocytes from neither mice nor rhesus macaques present CAR. Excess Ad5 fiber protein or anti-CAR antibody inhibits the binding of Ad5 to human erythrocytes and cryo-electron microscopy shows attachment via the fiber protein of Ad5, leading to close juxtaposition with the erythrocyte membrane. Human, but not murine, erythrocytes also present complement receptor (CR1), which binds Ad5 in the presence of antibodies and complement. Transplantation of human erythrocytes into nonobese diabetic/severe combined immunodeficiency mice extends blood circulation of intravenous Ad5 but decreases its extravasation into human xenograft tumors. Ad5 also shows extended circulation in transgenic mice presenting CAR on their erythrocytes, although it clears rapidly in transgenic mice presenting erythrocyte CR1. Hepatic infection is inhibited in both transgenic models. Erythrocytes may therefore restrict Ad5 infection (natural and therapeutic) in humans, independent of antibody status, presenting a formidable challenge to Ad5 therapeutics. "Stealthing" of Ad5 using hydrophilic polymers may enable circumvention of these natural virus traps.
Collapse
MESH Headings
- Adenovirus Infections, Human/blood
- Adenovirus Infections, Human/immunology
- Adenoviruses, Human/immunology
- Adenoviruses, Human/metabolism
- Adenoviruses, Human/physiology
- Animals
- Antigen Presentation/immunology
- Antigen Presentation/physiology
- Binding Sites
- Coxsackie and Adenovirus Receptor-Like Membrane Protein
- Erythrocytes/immunology
- Erythrocytes/metabolism
- Erythrocytes/virology
- Female
- HT29 Cells
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Models, Biological
- Receptors, Complement/immunology
- Receptors, Virus/immunology
- Tumor Cells, Cultured
- Virus Inactivation
Collapse
Affiliation(s)
- Robert C Carlisle
- Department of Clinical Pharmacology, University of Oxford, Oxford, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Miyazawa M, Yasuda M, Fujita M, Kajiwara H, Hirabayashi K, Takekoshi S, Hirasawa T, Murakami M, Ogane N, Kiguchi K, Ishiwata I, Mikami M, Osamura RY. Therapeutic strategy targeting the mTOR-HIF-1α-VEGF pathway in ovarian clear cell adenocarcinoma. Pathol Int 2009; 59:19-27. [DOI: 10.1111/j.1440-1827.2008.02320.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Bazan-Peregrino M, Carlisle RC, Hernandez-Alcoceba R, Iggo R, Homicsko K, Fisher KD, Halldén G, Mautner V, Shen Y, Seymour LW. Comparison of molecular strategies for breast cancer virotherapy using oncolytic adenovirus. Hum Gene Ther 2008; 19:873-86. [PMID: 18710328 DOI: 10.1089/hum.2008.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Oncolytic viruses are regulated by the tumor phenotype to replicate and lyse cancer cells selectively. To identify optimal strategies for breast cancer we compared five adenoviruses with distinct regulatory mechanisms: Ad-dl922-947 (targets G1-S checkpoint); Ad-Onyx-015 and Ad-Onyx-017 (target p53/mRNA export); Ad-vKH1 (targets Wnt pathway), and AdEHE2F (targets estrogen receptor/G1-S checkpoint/hypoxic signaling). The quantity of virus required to kill 50% of breast cancer cells after 6 days (EC(50), plaque-forming units per cell) was measured. The most potent virus was Ad-dl922-947 (EC(50), 0.01-5.4 in SkBr3, MDA-231, MDA-468, MCF7, and ZR75.1 cells), followed by wild-type (Ad-WT; EC(50), 0.3-5.5) and AdEHE2F (EC(50), 1.4-3.9). Ad-vKH1 (EC(50), 7.2-72.1), Ad-Onyx-017 (EC(50), 8.4-167), and Ad-Onyx-015 (EC(50), 17.7-377) showed less activity. Most viruses showed limited cytotoxicity in normal human cells, including breast epithelium MCF10A (EC(50), >722) and fibroblasts (EC(50), >192) and only moderate cytotoxicity in normal microvascular endothelial cells (HMVECs; EC(50), 42.8-149), except Ad-dl922-947, which was active in HMVECs (EC(50), 1.6). After injection into MDA-231 xenografts, Ad-WT, AdEHE2F, and Ad-dl922-947 showed replication, assessed by hexon staining and quantitative polymerase chain reaction measurement of viral DNA, and significantly inhibited tumor growth, leading to extended survival. After intravenous injection Ad-dl922-947 showed DNA replication (233% of the injected dose was measured in liver after 3 days) whereas AdEHE2F did not. Overall, AdEHE2F showed the best combination of low toxicity in normal cells and high activity in breast cancer in vitro and in vivo, suggesting that molecular targeting using estrogen response elements, hypoxia response elements, and a dysregulated G1-S checkpoint is a promising strategy for virotherapy of breast cancer.
Collapse
Affiliation(s)
- M Bazan-Peregrino
- Department of Clinical Pharmacology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Peerlinck I, Amini-Nik S, Phillips RK, Iggo R, Lemoine NR, Tejpar S, Vassaux G. Therapeutic potential of replication-selective oncolytic adenoviruses on cells from familial and sporadic desmoid tumors. Clin Cancer Res 2008; 14:6187-92. [PMID: 18829497 DOI: 10.1158/1078-0432.ccr-08-0410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Constitutive activation of the Wnt signaling pathway is a hallmark of many cancers and has been associated with familial and sporadic desmoid tumors. The aim of the present study is to assess the therapeutic potential of oncolytic adenoviruses selectively replicating in cells in which the Wnt signaling pathway is active on primary cells from desmoid tumors. EXPERIMENTAL DESIGN Primary cells extracted from familial (n = 3) or sporadic (n = 3) desmoid tumors were cultured short term. Cancer cell survival and viral replication were measured in vitro upon infection with two different oncolytic adenoviruses targeting a constitutive activation of the Wnt signaling pathway. Adenoviral infectivity was also assessed. RESULTS Although cells extracted from one sporadic desmoid tumor responded very well to the oncolytic action of the adenoviruses (<20% of viable cells upon infection at a multiplicity of infection of 10), cells from two tumor samples were totally resistant to the viral action. Cells from the remaining samples showed intermediate sensitivity to the oncolytic viruses. These effects were correlated to the level of infectivity of the cells. Finally, in responder cells, evidences of viral replication was observed. CONCLUSIONS Our experimental data suggest that the response of desmoid tumor cells to oncolytic adenovirus is neither correlated to the type of mutation activating the Wnt signaling pathway nor to the familial or sporadic nature of the tumor. In addition, they highlight the variability of infectivity of individual tumors and predict a great variability in the response to oncolytic adenoviruses.
Collapse
Affiliation(s)
- Inge Peerlinck
- Institute of Cancer and the CR-UK Clinical Centre, Barts and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
40
|
Huynh H, Chow KHP, Soo KC, Toh HC, Choo SP, Foo KF, Poon D, Ngo VC, Tran E. RAD001 (everolimus) inhibits tumour growth in xenograft models of human hepatocellular carcinoma. J Cell Mol Med 2008; 13:1371-80. [PMID: 18466352 PMCID: PMC4496150 DOI: 10.1111/j.1582-4934.2008.00364.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common malignancy worldwide and highly resistant to available chemotherapies. Mammalian target of rapamycin (mTOR) functions to regulate protein translation, angiogenesis and cell cycle progression in many cancers including HCC. In the present study, subcutaneous patient-derived HCC xenografts were used to study the effects of an mTOR inhibitor, RAD001 (everolimus), on tumour growth, apoptosis and angiogenesis. We report that oral administration of RAD001 to mice bearing patient-derived HCC xenografts resulted in a dose-dependent inhibition of tumour growth. RAD001-induced growth suppression was associated with inactivation of downstream targets of mTOR, reduction in VEGF expression and microvessel density, inhibition of cell proliferation, up-regulation of p27(Kip1) and down-regulation of p21(Cip1/Waf1), Cdk-6, Cdk-2, Cdk-4, cdc-25C, cyclin B1 and c-Myc. Our data indicate that the mTOR pathway plays an important role in angiogenesis, cell cycle progression and proliferation of liver cancer cells. Our study provides a strong rationale for clinical investigation of mTOR inhibitor RAD001 in patients with HCC.
Collapse
Affiliation(s)
- Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Funston GM, Kallioinen SE, de Felipe P, Ryan MD, Iggo RD. Expression of heterologous genes in oncolytic adenoviruses using picornaviral 2A sequences that trigger ribosome skipping. J Gen Virol 2008; 89:389-396. [PMID: 18198369 DOI: 10.1099/vir.0.83444-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Insertion of picornaviral 2A sequences into mRNAs causes ribosomes to skip formation of a peptide bond at the junction of the 2A and downstream sequences, leading to the production of two proteins from a single open reading frame. Adenoviral protein IX is a minor capsid protein that has been used to display foreign peptides on the surface of the capsid. We have used 2A sequences from the foot-and-mouth disease virus (FMDV) and porcine teschovirus 1 (PTV-1) to express protein IX (pIX) and green fluorescent protein (GFP) from pIX-2A-GFP fusion genes in an oncolytic virus derived from human adenovirus 5. GFP was efficiently expressed by constructs containing either 2A sequence. Peptide bond skipping was more efficient with the 58 aa FMDV sequence than with the 22 aa PTV-1 2A sequence, but the virus with the FMDV 2A sequence showed a reduction in plaque size, cytopathic effect, viral burst size and capsid stability. We conclude that ribosome skipping induced by 2A sequences is an effective strategy to express heterologous genes in adenoviruses; however, careful selection or optimization of the 2A sequence may be required if protein IX is used as the fusion partner.
Collapse
Affiliation(s)
- Garth M Funston
- School of Medicine, Biomolecular Sciences Building, University of St Andrews, St Andrews KY16 9ST, UK
| | - Susanna E Kallioinen
- School of Medicine, Biomolecular Sciences Building, University of St Andrews, St Andrews KY16 9ST, UK
| | - Pablo de Felipe
- School of Medicine, Biomolecular Sciences Building, University of St Andrews, St Andrews KY16 9ST, UK
| | - Martin D Ryan
- School of Biology, Biomolecular Sciences Building, University of St Andrews, St Andrews KY16 9ST, UK
| | - Richard D Iggo
- School of Medicine, Biomolecular Sciences Building, University of St Andrews, St Andrews KY16 9ST, UK
| |
Collapse
|
42
|
Delta-24-RGD in combination with RAD001 induces enhanced anti-glioma effect via autophagic cell death. Mol Ther 2008; 16:487-93. [PMID: 18253154 DOI: 10.1038/sj.mt.6300400] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Novel therapies are clearly needed for the treatment of gliomas, and strategies that involve combining oncolytic vectors with chemotherapy hold out significant hope for a more effective treatment of this malignancy. Whether chemotherapy acts directly on tumor cells by inducing cell arrest or cell death, or indirectly by blocking tumor angiogenesis, the resulting delay in tumor growth may provide the oncolytic virus with a wider window of opportunity to overcome the challenge imposed by the growth kinetics of the tumor. In this study we sought to determine whether the oncolytic adenovirus Delta-24-RGD, in combination with everolimus (RAD001), would result in an enhanced anti-glioma effect in vivo. Viability assays showed that Delta-24-RGD antitumoral activity is synergistically enhanced by combination with RAD001. Interestingly, combination treatment of Delta-24-RGD with RAD001 induced autophagy in vitro. We showed that Delta-24-RGD improved survival of tumor-bearing animals in a dose-dependent manner. A significant finding was that RAD001 enhanced the anti-glioma effect of Delta-24-RGD and resulted in the long-term survival of 80% of the experimental animals. Immunostaining of the treated tumors showed upregulation of Atg5, thereby indicating the therapeutic induction of autophagy. This is the first report showing that Delta-24-RGD plus RAD001 causes autophagic cell death, and dramatically increases long-term survival rates of glioma-bearing animals.
Collapse
|
43
|
LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat 2008; 11:32-50. [PMID: 18166498 PMCID: PMC2442829 DOI: 10.1016/j.drup.2007.11.003] [Citation(s) in RCA: 606] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 11/19/2007] [Accepted: 11/19/2007] [Indexed: 12/15/2022]
Abstract
The PI3K/Akt/mTOR pathway is a prototypic survival pathway that is constitutively activated in many types of cancer. Mechanisms for pathway activation include loss of tumor suppressor PTEN function, amplification or mutation of PI3K, amplification or mutation of Akt, activation of growth factor receptors, and exposure to carcinogens. Once activated, signaling through Akt can be propagated to a diverse array of substrates, including mTOR, a key regulator of protein translation. This pathway is an attractive therapeutic target in cancer because it serves as a convergence point for many growth stimuli, and through its downstream substrates, controls cellular processes that contribute to the initiation and maintenance of cancer. Moreover, activation of the Akt/mTOR pathway confers resistance to many types of cancer therapy, and is a poor prognostic factor for many types of cancers. This review will provide an update on the clinical progress of various agents that target the pathway, such as the Akt inhibitors perifosine and PX-866 and mTOR inhibitors (rapamycin, CCI-779, RAD-001) and discuss strategies to combine these pathway inhibitors with conventional chemotherapy, radiotherapy, as well as newer targeted agents. We will also discuss how the complex regulation of the PI3K/Akt/mTOR pathway poses practical issues concerning the design of clinical trials, potential toxicities and criteria for patient selection.
Collapse
Affiliation(s)
- Jaclyn LoPiccolo
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20889
| | - Gideon M. Blumenthal
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20889
| | - Wendy B. Bernstein
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20889
| | - Phillip A. Dennis
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20889
| |
Collapse
|
44
|
Stanford MM, Shaban M, Barrett JW, Werden SJ, Gilbert PA, Bondy-Denomy J, Mackenzie L, Graham KC, Chambers AF, McFadden G. Myxoma virus oncolysis of primary and metastatic B16F10 mouse tumors in vivo. Mol Ther 2007; 16:52-9. [PMID: 17998900 DOI: 10.1038/sj.mt.6300348] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Myxoma virus (MV) is a rabbit-specific poxvirus, whose unexpected tropism to human cancer cells has led to studies exploring its potential use in oncolytic therapy. MV infects a wide range of human cancer cells in vitro, in a manner intricately linked to the cellular activation of Akt kinase. MV has also been successfully used for treating human glioma xenografts in immunodeficient mice. This study examines the effectiveness of MV in treating primary and metastatic mouse tumors in immunocompetent C57BL6 mice. We have found that several mouse tumor cell lines, including B16 melanomas, are permissive to MV infection. B16F10 cells were used for assessing MV replication and efficacy in syngeneic primary tumor and metastatic models in vivo. Multiple intratumoral injections of MV resulted in dramatic inhibition of tumor growth. Systemic administration of MV in a lung metastasis model with B16F10LacZ cells was dramatically effective in reducing lung tumor burden. Combination therapy of MV with rapamycin reduced both size and number of lung metastases, and also reduced the induced antiviral neutralizing antibody titres, but did not affect tumor tropism. These results show MV to be a promising virotherapeutic agent in immunocompetent animal tumor models, with good efficacy in combination with rapamycin.
Collapse
Affiliation(s)
- Marianne M Stanford
- BioTherapeutics Research Group, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Merron A, Peerlinck I, Martin-Duque P, Burnet J, Quintanilla M, Mather S, Hingorani M, Harrington K, Iggo R, Vassaux G. SPECT/CT imaging of oncolytic adenovirus propagation in tumours in vivo using the Na/I symporter as a reporter gene. Gene Ther 2007; 14:1731-8. [PMID: 17960161 DOI: 10.1038/sj.gt.3303043] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Oncolytic adenoviruses have shown some promise in cancer gene therapy. However, their efficacy in clinical trials is often limited, and additional therapeutic interventions have been proposed to increase their efficacies. In this context, molecular imaging of viral spread in tumours could provide unique information to rationalize the timing of these combinations. Here, we use the human sodium iodide symporter (hNIS) as a reporter gene in wild-type and replication-selective adenoviruses. By design, hNIS cDNA is positioned in the E3 region in a wild-type adenovirus type 5 (AdIP1) and in an adenovirus in which a promoter from the human telomerase gene (RNA component) drives E1 expression (AdAM6). Viruses show functional hNIS expression and replication in vitro and kinetics of spread of the different viruses in tumour xenografts are visualized in vivo using a small animal nano-SPECT/CT camera. The time required to reach maximal spread is 48 h for AdIP1 and 72 h for AdAM6 suggesting that genetic engineering of adenoviruses can affect their kinetics of spread in tumours. Considering that this methodology is potentially clinically applicable, we conclude that hNIS-mediated imaging of viral spread in tumours may be an important tool for combined anticancer therapies involving replicating adenoviruses
Collapse
Affiliation(s)
- A Merron
- Centre for Molecular Oncology, Institute of Cancer, Queen Mary's School of Medicine and Dentistry, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tuncyurek P, Mayer JM, Klug F, Dillmann S, Henne-Bruns D, Keller F, Stracke S. Everolimus and mycophenolate mofetil sensitize human pancreatic cancer cells to gemcitabine in vitro: a novel adjunct to standard chemotherapy? Eur Surg Res 2007; 39:380-7. [PMID: 17700025 DOI: 10.1159/000107356] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Accepted: 06/06/2007] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIMS Gemcitabine improves survival in pancreatic adenocarcinoma. A variety of drugs have been tested to potentiate gemcitabine treatment for pancreatic cancer cells. Two major immunosuppressive drugs, mycophenolate mofetil (MMF) and everolimus (RAD001) have been shown to exert an anti-tumoral effect, but their ability to sensitize human pancreatic cell lines during gemcitabine treatment remains unclear. We examined the effects of everolimus and MMF on gemcitabine-treated MiaPaCa and Panc-1 cell lines. METHODS MiaPaCa and Panc-1 human pancreatic tumor cell lines were subjected to everolimus (0.001-1 microg/ml) or MMF (0.1-100 microg/ml) treatment in combination with gemcitabine (1-10(6) nM). Western blot analysis was performed for Panc-1 cells in the presence or absence of TGF-beta1 and different treatments: 0.1-100 muicro/ml MMF and 0.1-100 microg/ml everolimus. The antiproliferative effect of the treatment was assessed by BrdU test. The results were evaluated by two-way analysis of variance followed by post-hoc tests, and nonlinear regression analysis for dose-response rates. RESULTS As expected, standard treatment doses of gemcitabine decreased proliferation dose-dependently. Everolimus increased the actual EC(50) response to gemcitabine treatment (1-10(3) nM) to as much as 83.1 and 82.1% in MiaPaCa and Panc-1 cell lines, respectively. Likewise, concomitant administration with MMF altered the EC(50) of gemcitabine treatment in MiaPaCa cell lines to values between 76.8 and 85.2% for doses of >or=1 microg/ml. Even the minor dose of MMF (0.1 microg/ml) increased the antiproliferative effect of gemcitabine by 43.5% for MiaPaCa and 42.4% for Panc-1 cells. In addition, treatment of Panc-1 cells with MMF (0.1-100 microg/ml) dose-dependently inhibited TGF-beta1-induced collagen expression. CONCLUSION We found an overadditive antiproliferative effect of both MMF and everolimus in gemcitabine-treated MiaPaCa and Panc-1 cells in vitro, and an additional inhibitory effect of MMF on TGF-beta1-induced collagen type I expression. Interestingly, both the sensitizing effect of pancreatic cancer cells to gemcitabine treatment and the inhibition of collagen type I expression could be achieved by clinically feasible doses of everolimus and MMF. The use of these drugs is promising as a novel adjunct to standard chemotherapy.
Collapse
Affiliation(s)
- P Tuncyurek
- Department of Surgery, Adnan Menderes University, Aydin, Turkey
| | | | | | | | | | | | | |
Collapse
|
47
|
Alonso MM, Gomez-Manzano C, Jiang H, Bekele NB, Piao Y, Yung WKA, Alemany R, Fueyo J. Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo. Cancer Gene Ther 2007; 14:756-61. [PMID: 17557108 DOI: 10.1038/sj.cgt.7701067] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Novel therapies are clearly needed for gliomas, and the combination of oncolytic vectors with chemotherapy possesses a significant hope for the treatment of this malignancy. In addition, combination with chemotherapy allows for lower virus doses to achieve anticancer effect, thus resulting in lower undesirable toxicities due to viral proteins. In this work, we sought to determine whether combination of an oncolytic adenovirus ICOVIR-5, with RAD001 or temozolomide (TMZ) could result in enhanced anti-glioma effect in vivo. We assessed the in vitro cytotoxic effect and replication properties of ICOVIR-5 in combination with RAD001 or TMZ in U87 MG glioma cell line by MTT and TCID(50), respectively. Our data showed that in vitro treatment with RAD001 or TMZ not only interfered with adenovirus replication but, in addition, enhanced its oncolytic properties. To evaluate the in vivo anticancer effect, athymic mice bearing glioma xenografts (5 x 10(5) U87 MG cells/animal) received a single intratumoral injection of ICOVIR-5 (10(7) PFU/animal). RAD001 was given as a regimen of 5 mg/kg 5 days per week until the end of the experiment and TMZ was administered for 5 days at 7.5 mg/kg/mice. Of significance, combination of ICOVIR-5 with RAD001 or TMZ showed a potent anti-glioma effect in vivo, resulting in a dramatic extension of the median animal survival and in 20-40% animals becoming free of disease beyond 90 days.
Collapse
Affiliation(s)
- M M Alonso
- Department of Neuro-oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kuroda T, Rabkin SD, Martuza RL. Effective Treatment of Tumors with Strong β-Catenin/T-Cell Factor Activity by Transcriptionally Targeted Oncolytic Herpes Simplex Virus Vector. Cancer Res 2006; 66:10127-35. [PMID: 17047077 DOI: 10.1158/0008-5472.can-06-2744] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Wnt/beta-catenin/T-cell factor (Tcf) pathway is aberrantly up-regulated in the majority of colorectal cancers (CRC) and hepatoblastomas due to either an APC or beta-catenin gene mutation. We constructed synthetic promoters, T and TE, which contain tandem repeats of a Tcf responsive element without and with the human 4F2 gene intronic enhancer, respectively. Although the T and TE promoters showed higher transcriptional activity than a control promoter in all CRC and hepatoblastoma cell lines tested, with low activities in most other tumor cell lines, the level of transcription varied considerably among the CRC and hepatoblastoma cell lines. In some CRC cell lines, the TE promoter displayed higher levels of transcription than even the human CMV(IE) promoter. In those CRC cells, the APC gene mutations were located within a small segment between the first and second 20-amino-acid repeats in the mutation cluster region of the APC protein. We created a transcriptionally targeted oncolytic herpes simplex virus vector (bM24-TE) in which replication is driven by the TE promoter. This vector efficiently and specifically replicated in and killed tumor cells with strong beta-catenin/Tcf signaling. Intratumoral injection of bM24-TE significantly reduced the growth of highly beta-catenin active SW480 CRC tumors and induced a complete response in half of them, whereas it had no effect on the growth of beta-catenin-inactive A549 tumors. Our results suggest that a transcriptionally regulated oncolytic herpes vector targeting beta-catenin/Tcf signal is very efficacious against CRC tumors carrying an APC gene mutation between the first and second 20-amino-acid repeats.
Collapse
Affiliation(s)
- Toshihiko Kuroda
- Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | |
Collapse
|
49
|
Abstract
The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that controls many aspects of cellular physiology, including transcription, translation, cell size, cytoskeletal organization and autophagy. Recent advances in the mTOR signaling field have found that mTOR exists in two heteromeric complexes, mTORC1 and mTORC2. The activity of mTORC1 is regulated by the integration of many signals, including growth factors, insulin, nutrients, energy availability and cellular stressors such as hypoxia, osmotic stress, reactive oxygen species and viral infection. In this review we highlight recent advances in the mTOR signaling field that relate to how the two mTOR complexes are regulated, and we discuss stress conditions linked to the mTOR signaling network that have not been extensively covered in other reviews. Given the diversity of signals that have been shown to impinge on mTOR, we also speculate on other signal-transduction pathways that may be linked to mTOR in the future.
Collapse
Affiliation(s)
- M N Corradetti
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
50
|
Fuerer C, Homicsko K, Lukashev AN, Pittet AL, Iggo RD. Fusion of the BCL9 HD2 domain to E1A increases the cytopathic effect of an oncolytic adenovirus that targets colon cancer cells. BMC Cancer 2006; 6:236. [PMID: 17020613 PMCID: PMC1609183 DOI: 10.1186/1471-2407-6-236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Accepted: 10/04/2006] [Indexed: 12/20/2022] Open
Abstract
Background The Wnt signaling pathway is activated by mutations in the APC and β-catenin genes in many types of human cancer. β-catenin is stabilized by these mutations and activates transcription in part by acting as a bridge between Tcf/LEF proteins and the HD2 domain of the BCL9 coactivator. We have previously described oncolytic adenoviruses with binding sites for Tcf/LEF transcription factors inserted into the early viral promoters. These viruses replicate selectively in cells with activation of the Wnt pathway. To increase the activity of these viruses we have fused the viral transactivator E1A to the BCL9 HD2 domain. Methods Luciferase assays, co-immunoprecipitation and Western blotting, immunofluorescent cell staining and cytopathic effect assays were used to characterize the E1A-HD2 fusion protein and virus in vitro. Growth curves of subcutaneous SW620 colon cancer xenografts were used to characterize the virus in vivo. Results The E1A-HD2 fusion protein binds to β-catenin in vivo and activates a Tcf-regulated luciferase reporter better than wild-type E1A in cells with activated Wnt signaling. Expression of the E1A-HD2 protein promotes nuclear import of β-catenin, mediated by the strong nuclear localization signal in E1A. Tcf-regulated viruses expressing the fusion protein show increased expression of viral proteins and a five-fold increase in cytopathic effect (CPE) in colorectal cancer cell lines. There was no change in viral protein expression or CPE in HeLa cells, indicating that E1A-HD2 viruses retain selectivity for cells with activation of the Wnt signaling pathway. Despite increasing the cytopathic effect of the virus in vitro, fusion of the HD2 domain to E1A did not increase the burst size of the virus in vitro or the anti-tumor effect of the virus in an SW620 xenograft model in vivo. Conclusion Despite an increase in the nuclear pool of β-catenin, the effects on viral activity in colon cancer cells were small, suggesting that factors acting downstream of β-catenin are limiting for viral replication and toxicity in these cells. The approach of fusing E1A to a protein domain implicated in oncogenic signaling could be used to selectively increase the activity of oncolytic viruses targeting several other pathways defective in cancer.
Collapse
Affiliation(s)
- Christophe Fuerer
- NCCR Molecular Oncology Programme, Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland
| | - Krisztian Homicsko
- NCCR Molecular Oncology Programme, Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland
| | - Alexander N Lukashev
- NCCR Molecular Oncology Programme, Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland
| | - Anne-Laure Pittet
- NCCR Molecular Oncology Programme, Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland
| | - Richard D Iggo
- NCCR Molecular Oncology Programme, Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland
- Current address: Bute Medical School, University of St Andrews, Fife KY16 9TS, Scotland, UK
| |
Collapse
|