1
|
Gazal S, Gazal S, Kaur P, Bhan A, Olagnier D. Breaking Barriers: Animal viruses as oncolytic and immunotherapeutic agents for human cancers. Virology 2024; 600:110238. [PMID: 39293238 DOI: 10.1016/j.virol.2024.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
Oncolytic viruses, defined as viruses capable of lysing cancer cells, emerged as a groundbreaking class of therapeutic entities poised to revolutionize cancer treatment. Their mode of action encompasses both direct tumor cell lysis and the indirect enhancement of anti-tumor immune responses. Notably, four leading contenders in this domain, Rigvir® in Latvia, T-VEC in the United States, H101 in China and Teserpaturev (DELYTACT®) in Japan, have earned approval for treating metastatic melanoma (Rigvir and T-VEC), nasopharyngeal carcinoma and malignant glioma, respectively. Despite these notable advancements, the integration of oncolytic viruses into cancer therapy encounters several challenges. Foremost among these hurdles is the considerable variability observed in clinical responses to oncolytic virus interventions. Moreover, the adaptive immune system may inadvertently target the oncolytic viruses themselves, diverting immune resources away from tumor antigens and undermining therapeutic efficacy. Another significant limitation arises from the presence of preexisting immunity against oncolytic viruses in certain patient populations, hampering treatment outcomes. To circumvent this obstacle, researchers are investigating the utilization of animal viruses, for which humans lack preexisting immunity, as a compelling alternative to human-derived counterparts. In our comprehensive review, we delve into the intricate nuances of oncolytic virotherapy, elucidating the multifaceted mechanisms through which these viruses exert their anti-cancer effects. Furthermore, we provide a thorough examination of animal-derived oncolytic viruses, highlighting their respective strengths and limitations. Lastly, we explore the promising potential of leveraging animal viruses as potent oncolytic agents, offering new avenues for enhancing the efficacy and reach of human cancer therapeutics.
Collapse
Affiliation(s)
- Sabahat Gazal
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu, Jammu & Kashmir, India; Department of Biomedicine, Aarhus University, Denmark
| | - Sundus Gazal
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu, Jammu & Kashmir, India.
| | - Paviter Kaur
- Division of Veterinary Microbiology, College of Veterinary Sciences, Guru Angad Dev Veterinary and Animal Science University, Ludhiana, Punjab, India
| | - Anvesha Bhan
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu, Jammu & Kashmir, India
| | | |
Collapse
|
2
|
Gujar S, Pol JG, Kumar V, Lizarralde-Guerrero M, Konda P, Kroemer G, Bell JC. Tutorial: design, production and testing of oncolytic viruses for cancer immunotherapy. Nat Protoc 2024; 19:2540-2570. [PMID: 38769145 DOI: 10.1038/s41596-024-00985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 02/12/2024] [Indexed: 05/22/2024]
Abstract
Oncolytic viruses (OVs) represent a novel class of cancer immunotherapy agents that preferentially infect and kill cancer cells and promote protective antitumor immunity. Furthermore, OVs can be used in combination with established or upcoming immunotherapeutic agents, especially immune checkpoint inhibitors, to efficiently target a wide range of malignancies. The development of OV-based therapy involves three major steps before clinical evaluation: design, production and preclinical testing. OVs can be designed as natural or engineered strains and subsequently selected for their ability to kill a broad spectrum of cancer cells rather than normal, healthy cells. OV selection is further influenced by multiple factors, such as the availability of a specific viral platform, cancer cell permissivity, the need for genetic engineering to render the virus non-pathogenic and/or more effective and logistical considerations around the use of OVs within the laboratory or clinical setting. Selected OVs are then produced and tested for their anticancer potential by using syngeneic, xenograft or humanized preclinical models wherein immunocompromised and immunocompetent setups are used to elucidate their direct oncolytic ability as well as indirect immunotherapeutic potential in vivo. Finally, OVs demonstrating the desired anticancer potential progress toward translation in patients with cancer. This tutorial provides guidelines for the design, production and preclinical testing of OVs, emphasizing considerations specific to OV technology that determine their clinical utility as cancer immunotherapy agents.
Collapse
Affiliation(s)
- Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Jonathan G Pol
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Vishnupriyan Kumar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Manuela Lizarralde-Guerrero
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
- Ecole Normale Supérieure de Lyon, Lyon, France
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Guido Kroemer
- INSERM, U1138, Paris, France.
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
- Université Paris Cité, Paris, France.
- Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Institut Universitaire de France, Paris, France.
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - John C Bell
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada.
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
3
|
Al-Shammari AM, Salman MI. Antimetastatic and antitumor activities of oncolytic NDV AMHA1 in a 3D culture model of breast cancer. Front Mol Biosci 2024; 11:1331369. [PMID: 39281317 PMCID: PMC11392722 DOI: 10.3389/fmolb.2024.1331369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/29/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Newcastle disease virus (NDV) AMHA1 is capable of killing cancer cells by direct replication or induction of apoptosis alongside other pathways. In this study, we report the potent antimetastatic and anticancer activities of NDV AMHA1 in a 3D spheroid model of breast cancer metastasis. Methods we used two breast cancer cell lines AMJ13 and MCF7 in our metastasis model system. Results First, we showed that NDV AMHA1 can infect and kill breast cancer cells in proliferating adherent cells and tumor spheroids using different virus doses and studying virus replication kinetics. We showed that NDV can infect and spread within the spheroids that represent metastasis before and after reattachment. Furthermore, we evaluated the ability of NDV to induce apoptosis in cancer spheroids and by virus tracking showed that NDV infection is essential for the elimination of these metastasis spheroids. Discussion The mechanism by which NDV induces cell killing in the metastasis model is the induction of caspase-3 and P21 and inhibition of Ki67 in cancer cells, but not in normal cells. In conclusion, these results indicate that NDV AMHA1 has the ability to kill breast cancer metastases in suspension or attached, and this is a novel finding of NDV AMHA1 being a possibly efficient therapy against human metastatic breast cancer.
Collapse
Affiliation(s)
- Ahmed Majeed Al-Shammari
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| | - Marwa Ibrahim Salman
- Department of Biotechnology, College of Science, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
4
|
Rahman MM, Wells G, Rantala JK, Helleday T, Muthana M, Danson SJ. In-vitro assays for immuno-oncology drug efficacy assessment and screening for personalized cancer therapy: scopes and challenges. Expert Rev Clin Immunol 2024; 20:821-838. [PMID: 38546609 DOI: 10.1080/1744666x.2024.2336583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Immunotherapies have revolutionized cancer treatment, but often fail to produce desirable therapeutic outcomes in all patients. Due to the inter-patient heterogeneity and complexity of the tumor microenvironment, personalized treatment approaches are gaining demand. Researchers have long been using a range of in-vitro assays including 2D models, organoid co-cultures, and cancer-on-a-chip platforms for cancer drug screening. A comparative analysis of these assays with their suitability, high-throughput capacity, and clinical translatability is required for optimal translational use. AREAS COVERED The review summarized in-vitro platforms with their comparative advantages and limitations including construction strategies, and translational potential for immuno-oncology drug efficacy assessment. We also discussed end-point analysis strategies so that researchers can contextualize their usefulness and optimally design experiments for personalized immunotherapy efficacy prediction. EXPERT OPINION Researchers developed several in-vitro platforms that can provide information on personalized immunotherapy efficacy from different angles. Image-based assays are undoubtedly more suitable to gather a wide range of information including cellular morphology and phenotypical behaviors but need significant improvement to overcome issues including background noise, sample preparation difficulty, and long duration of experiment. More studies and clinical trials are needed to resolve these issues and validate the assays before they can be used in real-life scenarios.
Collapse
Affiliation(s)
- Md Marufur Rahman
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Directorate General of Health Services, Dhaka, Bangladesh
| | - Greg Wells
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Juha K Rantala
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Misvik Biology Ltd, Turku, Finland
| | - Thomas Helleday
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Department of Oncology-Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Munitta Muthana
- Nanobug Oncology Sheffield, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Sarah J Danson
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
5
|
Marquette CA, Petiot E, Spindler A, Ebel C, Nzepa M, Moreau B, Erbs P, Balloul JM, Quemeneur E, Zaupa C. 3D bioprinted CRC model brings to light the replication necessity of an oncolytic vaccinia virus encoding FCU1 gene to exert an efficient anti-tumoral activity. Front Oncol 2024; 14:1384499. [PMID: 39091906 PMCID: PMC11292208 DOI: 10.3389/fonc.2024.1384499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
The oncolytic virus represents a promising therapeutic strategy involving the targeted replication of viruses to eliminate cancer cells, while preserving healthy ones. Despite ongoing clinical trials, this approach encounters significant challenges. This study delves into the interaction between an oncolytic virus and extracellular matrix mimics (ECM mimics). A three-dimensional colorectal cancer model, enriched with ECM mimics through bioprinting, was subjected to infection by an oncolytic virus derived from the vaccinia virus (oVV). The investigation revealed prolonged expression and sustained oVV production. However, the absence of a significant antitumor effect suggested that the virus's progression toward non-infected tumoral clusters was hindered by the ECM mimics. Effective elimination of tumoral cells was achieved by introducing an oVV expressing FCU1 (an enzyme converting the prodrug 5-FC into the chemotherapeutic compound 5-FU) alongside 5-FC. Notably, this efficacy was absent when using a non-replicative vaccinia virus expressing FCU1. Our findings underscore then the crucial role of oVV proliferation in a complex ECM mimics. Its proliferation facilitates payload expression and generates a bystander effect to eradicate tumors. Additionally, this study emphasizes the utility of 3D bioprinting for assessing ECM mimics impact on oVV and demonstrates how enhancing oVV capabilities allows overcoming these barriers. This showcases the potential of 3D bioprinting technology in designing purpose-fit models for such investigations.
Collapse
Affiliation(s)
- Christophe A. Marquette
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne, France
| | - Emma Petiot
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne, France
| | | | | | - Mael Nzepa
- Transgene SA, Illkirch-Graffenstaden, France
| | | | | | | | | | | |
Collapse
|
6
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [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: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Lauer UM, Beil J. Oncolytic viruses: challenges and considerations in an evolving clinical landscape. Future Oncol 2022; 18:2713-2732. [PMID: 35818970 DOI: 10.2217/fon-2022-0440] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite advances in treatment, cancer remains a leading cause of death worldwide. Although treatment strategies are continually progressing, cancers have evolved many mechanisms for evading therapies and the host immune system. Oncolytic viruses (OVs) could provide a much-needed option for cancers that are resistant to existing treatments. OVs can be engineered to specifically target and kill cancer cells, while simultaneously triggering an immune response at the site of infection. This review will focus on the challenges of developing a successful OV and translation to clinical practice, discussing the innovative strategies that are being used to optimize the potential of OVs. Here, we will also explore the current clinical landscape and the prospects of OVs in early clinical development.
Collapse
Affiliation(s)
- Ulrich M Lauer
- Department of Internal Medicine VIII, Virotherapy Center Tübingen, Medical Oncology & Pneumology, Medical University Hospital Tübingen, Otfried-Mueller-Str. 10, Tübingen, 72076, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, Otfried-Mueller-Str. 10, Tübingen, 72076, Germany
| | - Julia Beil
- Department of Internal Medicine VIII, Virotherapy Center Tübingen, Medical Oncology & Pneumology, Medical University Hospital Tübingen, Otfried-Mueller-Str. 10, Tübingen, 72076, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, Otfried-Mueller-Str. 10, Tübingen, 72076, Germany
| |
Collapse
|
8
|
Differential In Vitro Growth and Cell Killing of Cancer versus Benign Prostate Cells by Oncolytic Parainfluenza Virus. Pathogens 2022; 11:pathogens11050493. [PMID: 35631014 PMCID: PMC9147676 DOI: 10.3390/pathogens11050493] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
The development of effective oncolytic viruses will require understanding the differences in virus replication and killing between normal and cancer cells. Here, we have evaluated infections of metastatic cancer (22Rv1) and benign non-tumorigenic (BPH-1) prostate cell lines with a mutant parainfluenza virus 5 (P/V/F) encoding a defective V protein and a hyperfusogenic F protein. Under low multiplicity of infection (MOI), the P/V/F mutant efficiently spread in 22Rv1 cells but was restricted in BPH-1 cells due to type-I interferon (IFN-I) responses. In mixed co-cultures, the P/V/F mutant showed specificity towards and spread within the 22Rv1 cells versus BPH-1 cells. Under high MOI conditions, both BPH-1 and 22Rv1 cells showed efficient infection by the P/V/F mutant. However, compared to BPH-1 cells, the 22Rv1 cancer cells showed increased cytopathic effect, higher induction of caspase-8 and -9, and extensive syncytia formation. In 22Rv1 spheroid cultures, P/V/F infection was less efficient compared to monolayers, but the virus was able to spread through spheroids and induce death. These data indicate that IFN-I sensitivity is a major determinant of specificity of P/V/F spread through populations of cancer versus benign cells, and additionally, differences in activation of apoptotic pathways and syncytia formation can contribute to differential outcomes in cancer versus benign cells.
Collapse
|
9
|
Braccini S, Tacchini C, Chiellini F, Puppi D. Polymeric Hydrogels for In Vitro 3D Ovarian Cancer Modeling. Int J Mol Sci 2022; 23:3265. [PMID: 35328686 PMCID: PMC8954571 DOI: 10.3390/ijms23063265] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) grows and interacts constantly with a complex microenvironment, in which immune cells, fibroblasts, blood vessels, signal molecules and the extracellular matrix (ECM) coexist. This heterogeneous environment provides structural and biochemical support to the surrounding cells and undergoes constant and dynamic remodeling that actively promotes tumor initiation, progression, and metastasis. Despite the fact that traditional 2D cell culture systems have led to relevant medical advances in cancer research, 3D cell culture models could open new possibilities for the development of an in vitro tumor microenvironment more closely reproducing that observed in vivo. The implementation of materials science and technology into cancer research has enabled significant progress in the study of cancer progression and drug screening, through the development of polymeric scaffold-based 3D models closely recapitulating the physiopathological features of native tumor tissue. This article provides an overview of state-of-the-art in vitro tumor models with a particular focus on 3D OC cell culture in pre-clinical studies. The most representative OC models described in the literature are presented with a focus on hydrogel-based scaffolds, which guarantee soft tissue-like physical properties as well as a suitable 3D microenvironment for cell growth. Hydrogel-forming polymers of either natural or synthetic origin investigated in this context are described by highlighting their source of extraction, physical-chemical properties, and application for 3D ovarian cancer cell culture.
Collapse
Affiliation(s)
| | | | | | - Dario Puppi
- BioLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM-Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (S.B.); (C.T.)
| |
Collapse
|
10
|
Yee C, Dickson KA, Muntasir MN, Ma Y, Marsh DJ. Three-Dimensional Modelling of Ovarian Cancer: From Cell Lines to Organoids for Discovery and Personalized Medicine. Front Bioeng Biotechnol 2022; 10:836984. [PMID: 35223797 PMCID: PMC8866972 DOI: 10.3389/fbioe.2022.836984] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer has the highest mortality of all of the gynecological malignancies. There are several distinct histotypes of this malignancy characterized by specific molecular events and clinical behavior. These histotypes have differing responses to platinum-based drugs that have been the mainstay of therapy for ovarian cancer for decades. For histotypes that initially respond to a chemotherapeutic regime of carboplatin and paclitaxel such as high-grade serous ovarian cancer, the development of chemoresistance is common and underpins incurable disease. Recent discoveries have led to the clinical use of PARP (poly ADP ribose polymerase) inhibitors for ovarian cancers defective in homologous recombination repair, as well as the anti-angiogenic bevacizumab. While predictive molecular testing involving identification of a genomic scar and/or the presence of germline or somatic BRCA1 or BRCA2 mutation are in clinical use to inform the likely success of a PARP inhibitor, no similar tests are available to identify women likely to respond to bevacizumab. Functional tests to predict patient response to any drug are, in fact, essentially absent from clinical care. New drugs are needed to treat ovarian cancer. In this review, we discuss applications to address the currently unmet need of developing physiologically relevant in vitro and ex vivo models of ovarian cancer for fundamental discovery science, and personalized medicine approaches. Traditional two-dimensional (2D) in vitro cell culture of ovarian cancer lacks critical cell-to-cell interactions afforded by culture in three-dimensions. Additionally, modelling interactions with the tumor microenvironment, including the surface of organs in the peritoneal cavity that support metastatic growth of ovarian cancer, will improve the power of these models. Being able to reliably grow primary tumoroid cultures of ovarian cancer will improve the ability to recapitulate tumor heterogeneity. Three-dimensional (3D) modelling systems, from cell lines to organoid or tumoroid cultures, represent enhanced starting points from which improved translational outcomes for women with ovarian cancer will emerge.
Collapse
Affiliation(s)
- Christine Yee
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kristie-Ann Dickson
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Mohammed N. Muntasir
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Yue Ma
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Deborah J. Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
11
|
Varudkar N, Oyer JL, Copik A, Parks GD. Oncolytic parainfluenza virus combines with NK cells to mediate killing of infected and non-infected lung cancer cells within 3D spheroids: role of type I and type III interferon signaling. J Immunother Cancer 2021; 9:jitc-2021-002373. [PMID: 34172515 PMCID: PMC8237729 DOI: 10.1136/jitc-2021-002373] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND There is intense interest in developing novel oncolytic viruses, which can be used in cancer therapies along with immune cells such as natural killer (NK) cells. We have previously developed a particle-based method for in vitro expansion of highly cytotoxic human NK cells (PM21-NK cells). Here, we have tested the hypothesis that oncolytic parainfluenza virus 5 (P/V virus) can combine with PM21-NK cells for targeted killing of lung cancer cells. METHODS PM21-NK cells were assayed for killing of P/V virus-infected A549, H1299 and Calu-1 lung cancer cells in two-dimensional (2D) and three-dimensional (3D) cultures using flow cytometry, luminescence and kinetic imaging-based methods. Blocking antibodies were used to evaluate NK cell activating receptors involved in PM21-NK cell killing of infected target cells. Media transfer experiments tested soluble factors that increase PM21-NK cell killing of both P/V virus-infected and uninfected tumor cells. RESULTS In 2D cultures, PM21-NK cells efficiently killed P/V virus-infected cancer cells compared with non-infected cells, through involvement of the viral glycoprotein and NK cell receptors NKp30, NKp46 and NKG2D. In 3D spheroid cultures, P/V virus infection was restricted to the outer layer of the spheroid. However, PM21-NK cells were able to more efficiently kill both the outer layer of infected cells in the spheroid and progressing further to kill the uninfected interior cells. Media transfer experiments demonstrated that P/V virus infection produced both type I and type III interferons, which decreased cell growth, which contributed to a reduction in the overall number of uninfected tumor cells in conjunction with PM21-NK cells. Across five cancer cell lines, the contribution of P/V virus infection on PM21-NK cell killing of target cells correlated with interferon induction. CONCLUSION Our data support the potential of combining oncolytic parainfluenza virus with PM21-NK cell adoptive therapy against lung cancer.
Collapse
Affiliation(s)
- Namita Varudkar
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Jeremiah L Oyer
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Alicja Copik
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Griffith D Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
12
|
HIV-Infected Macrophages Are Infected and Killed by the Interferon-Sensitive Rhabdovirus MG1. J Virol 2021; 95:JVI.01953-20. [PMID: 33568507 PMCID: PMC8104113 DOI: 10.1128/jvi.01953-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) remains a treatable, but incurable, viral infection. The establishment of viral reservoirs containing latently infected cells remains the main obstacle in the search for a cure. The use of unique cell surface markers to target and eradicate HIV-infected cells has been a longstanding objective of HIV-1 cure research. This approach, however, overlooks the possibility that intracellular changes present within HIV-infected cells may serve as valuable therapeutic targets. For example, the identification of dysregulated antiviral signaling in cancer has led to the characterization of oncolytic viruses capable of preferentially killing cancer cells. Since impairment of cellular antiviral machinery has been proposed as a mechanism by which HIV-1 evades immune clearance, we hypothesized that HIV-infected macrophages (an important viral reservoir in vivo) would be preferentially killed by the interferon-sensitive oncolytic Maraba virus MG1. We first showed that HIV-infected monocyte-derived macrophages (MDM) were more susceptible to MG1 infection and killing than HIV-uninfected cells. As MG1 is highly sensitive to type I interferons (IFN-I), we then investigated whether we could identify IFN-I signaling differences between HIV-infected and uninfected MDM and found evidence of impaired IFN-α responsiveness within HIV-infected cells. Finally, to assess whether MG1 could target a relevant, primary cell reservoir of HIV-1, we investigated its effects in alveolar macrophages (AM) obtained from effectively treated individuals living with HIV-1. As observed with in vitro-infected MDM, we found that HIV-infected AM were preferentially eliminated by MG1. In summary, the oncolytic rhabdovirus MG1 appears to preferentially target and kill HIV-infected cells via impairment of antiviral signaling pathways and may therefore provide a novel approach to an HIV-1 cure. IMPORTANCE Human immunodeficiency virus type 1 (HIV-1) remains a treatable, but incurable, viral infection. The establishment of viral reservoirs containing latently infected cells remains the main obstacle in the search for a cure. Cure research has also focused on only one cellular target of HIV-1 (the CD4+ T cell) while largely overlooking others (such as macrophages) that contribute to HIV-1 persistence. In this study, we address these challenges by describing a potential strategy for the eradication of HIV-infected macrophages. Specifically, we show that an engineered rhabdovirus—initially developed as a cancer therapy—is capable of preferential infection and killing of HIV-infected macrophages, possibly via the same altered antiviral signaling seen in cancer cells. As this rhabdovirus is currently being explored in phase I/II clinical trials, there is potential for this approach to be readily adapted for use within the HIV-1 cure field.
Collapse
|
13
|
Wyżewski Z, Świtlik W, Mielcarska MB, Gregorczyk-Zboroch KP. The Role of Bcl-xL Protein in Viral Infections. Int J Mol Sci 2021; 22:ijms22041956. [PMID: 33669408 PMCID: PMC7920434 DOI: 10.3390/ijms22041956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/13/2021] [Accepted: 02/14/2021] [Indexed: 02/06/2023] Open
Abstract
Bcl-xL represents a family of proteins responsible for the regulation of the intrinsic apoptosis pathway. Due to its anti-apoptotic activity, Bcl-xL co-determines the viability of various virally infected cells. Their survival may determine the effectiveness of viral replication and spread, dynamics of systemic infection, and viral pathogenesis. In this paper, we have reviewed the role of Bcl-xL in the context of host infection by eight different RNA and DNA viruses: hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), influenza A virus (IAV), Epstein-Barr virus (EBV), human T-lymphotropic virus type-1 (HTLV-1), Maraba virus (MRBV), Schmallenberg virus (SBV) and coronavirus (CoV). We have described an influence of viral infection on the intracellular level of Bcl-xL and discussed the impact of Bcl-xL-dependent cell survival control on infection-accompanying pathogenic events such as tissue damage or oncogenesis. We have also presented anti-viral treatment strategies based on the pharmacological regulation of Bcl-xL expression or activity.
Collapse
Affiliation(s)
- Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University in Warsaw, 01-815 Warsaw, Poland
- Correspondence: ; Tel.: +48 728-208-338
| | - Weronika Świtlik
- Department of Biochemistry and Microbiology, Institute of Biology, Warsaw University of Life Sciences-SGGW, 02-787 Warsaw, Poland;
| | - Matylda Barbara Mielcarska
- Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (M.B.M.); (K.P.G.-Z.)
| | | |
Collapse
|
14
|
Fitzgerald SJ, Cobb JS, Janorkar AV. Comparison of the formation, adipogenic maturation, and retention of human adipose-derived stem cell spheroids in scaffold-free culture techniques. J Biomed Mater Res B Appl Biomater 2020; 108:3022-3032. [PMID: 32396702 PMCID: PMC8506838 DOI: 10.1002/jbm.b.34631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 11/02/2023]
Abstract
While three-dimensional spheroids outperform traditional two-dimensional monolayer culture for human adipose-derived stem cells (hASCs), there is not a consensus on the most successful method for enhancing their adipogenic differentiation and minimizing the loss of physiologically relevant, fatty spheroids during culture. To this end, we compared three culture methods, namely, elastin-like polypeptide-polyethyleneimine (ELP-PEI) coated surfaces, ultra-low attachment static culture, and suspension culture for their ability to form and retain productive hASC spheroids. The ELP-PEI coatings used the ELP conjugated to two molecular weights of PEI (800 or 25,000 g/mol). FTIR spectroscopy, atomic force microscopy, and contact angle goniometry revealed that the ELP-PEI coatings had similar chemical structures, surface topography, and hydrophobicity. Time-lapse microscopy showed that increasing the PEI molecular weight resulted in smaller spheroids. Measurement of triglyceride content showed that the three methods induced comparable differentiation of hASCs toward the adipogenic lineage. DNA content and morphometric analysis revealed merging of spheroids to form larger spheroids in the ultra-low attachment static culture and suspension culture methods. In contrast, the retention of hASC spheroid sizes and numbers with a regular spheroid size (~100 μm) were best atop the ELP-PEI800 coatings. Overall, this research shows that the spheroid culture atop the ELP-PEI coatings is a suitable cell culture model for future studies involving long-term, three-dimensional culture of mature adipocytes derived from hASCs.
Collapse
Affiliation(s)
- Sarah J. Fitzgerald
- Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216
| | - Jared S. Cobb
- Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216
| | - Amol V. Janorkar
- Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216
| |
Collapse
|
15
|
Rahman MM, McFadden G. Oncolytic Virotherapy with Myxoma Virus. J Clin Med 2020; 9:jcm9010171. [PMID: 31936317 PMCID: PMC7020043 DOI: 10.3390/jcm9010171] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/25/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses are one of the most promising novel therapeutics for malignant cancers. They selectively infect and kill cancer cells while sparing the normal counterparts, expose cancer- specific antigens and activate the host immune system against both viral and tumor determinants. Oncolytic viruses can be used as monotherapy or combined with existing cancer therapies to become more potent. Among the many types of oncolytic viruses that have been developed thus far, members of poxviruses are the most promising candidates against diverse cancer types. This review summarizes recent advances that are made with oncolytic myxoma virus (MYXV), a member of the Leporipoxvirus genus. Unlike other oncolytic viruses, MYXV infects only rabbits in nature and causes no harm to humans or any other non-leporid animals. However, MYXV can selectively infect and kill cancer cells originating from human, mouse and other host species. This selective cancer tropism and safety profile have led to the testing of MYXV in various types of preclinical cancer models. The next stage will be successful GMP manufacturing and clinical trials that will bring MYXV from bench to bedside for the treatment of currently intractable malignancies.
Collapse
|
16
|
Lee SW, Lee KJ, Jeong SY, Joo CH, Lee H, Jeong GS. Evaluation of Bystander Infection of Oncolytic Virus using a Medium Flow Integrated 3D In Vitro Microphysiological System. ACTA ACUST UNITED AC 2019; 4:e1900143. [DOI: 10.1002/adbi.201900143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/04/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Sang Woo Lee
- Biomedical Engineering Research CenterAsan Institute for Life ScienceAsan Medical Center Seoul 05505 Korea
| | - Kyoung Jin Lee
- Department of MicrobiologyUniversity of Ulsan College of Medicine Seoul 05505 Korea
- Bio‐Medical Institute of TechnologyAsan Medical Center Seoul 05505 Korea
| | - Soo Yeon Jeong
- Biomedical Engineering Research CenterAsan Institute for Life ScienceAsan Medical Center Seoul 05505 Korea
| | - Chul Hyun Joo
- Department of MicrobiologyUniversity of Ulsan College of Medicine Seoul 05505 Korea
- Bio‐Medical Institute of TechnologyAsan Medical Center Seoul 05505 Korea
| | - Heuiran Lee
- Department of MicrobiologyUniversity of Ulsan College of Medicine Seoul 05505 Korea
- Bio‐Medical Institute of TechnologyAsan Medical Center Seoul 05505 Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research CenterAsan Institute for Life ScienceAsan Medical Center Seoul 05505 Korea
- Department of Convergence MedicineUniversity of Ulsan College of Medicine Seoul 05505 Korea
| |
Collapse
|
17
|
Jayawardena N, Burga LN, Poirier JT, Bostina M. Virus-Receptor Interactions: Structural Insights For Oncolytic Virus Development. Oncolytic Virother 2019; 8:39-56. [PMID: 31754615 PMCID: PMC6825474 DOI: 10.2147/ov.s218494] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Recent advancements in oncolytic virotherapy commend a special attention to developing new strategies for targeting cancer cells with oncolytic viruses (OVs). Modifications of the viral envelope or coat proteins serve as a logical mean of repurposing viruses for cancer treatment. In this review, we discuss how detailed structural knowledge of the interactions between OVs and their natural receptors provide valuable insights into tumor specificity of some viruses and re-targeting of alternate receptors for broad tumor tropism or improved tumor selectivity.
Collapse
Affiliation(s)
- Nadishka Jayawardena
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Laura N Burga
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - John T Poirier
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Otago Micro and Nano Imaging, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
McGray AJR, Huang RY, Battaglia S, Eppolito C, Miliotto A, Stephenson KB, Lugade AA, Webster G, Lichty BD, Seshadri M, Kozbor D, Odunsi K. Oncolytic Maraba virus armed with tumor antigen boosts vaccine priming and reveals diverse therapeutic response patterns when combined with checkpoint blockade in ovarian cancer. J Immunother Cancer 2019; 7:189. [PMID: 31315674 PMCID: PMC6637574 DOI: 10.1186/s40425-019-0641-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cancer immunotherapies are emerging as promising treatment strategies for ovarian cancer patients that experience disease relapse following first line therapy. As such, identifying strategies to bolster anti-tumor immunity and limit immune suppression, while recognizing diverse patterns of tumor response to immunotherapy is critical to selecting treatment combinations that lead to durable therapeutic benefit. METHODS Using a pre-clinical mouse model, we evaluated a heterologous prime/boost vaccine in combination with checkpoint blockade to treat metastatic intraperitoneal ovarian cancer. Vaccine-elicited CD8+ T cell responses and changes in the tumor microenvironment following treatment were analyzed and compared to treatment outcome. Kinetics of intraperitoneal tumor growth were assessed using non-invasive magnetic resonance imaging (MRI). RESULTS Vaccine priming followed by antigen-armed oncolytic Maraba virus boosting elicited robust tumor-specific CD8+ T cell responses that improved tumor control and led to unique immunological changes in the tumor, including a signature that correlated with improved clinical outcome of ovarian cancer patients. However, this treatment was not curative and T cells in the tumor microenvironment (TME) were functionally suppressed. Combination PD-1 blockade partially overcame the adaptive resistance in the tumor observed in response to prime/boost vaccination, restoring CD8+ T cell function in the TME and enhancing the therapeutic response. Non-invasive MRI of tumors during the course of combination treatment revealed heterogeneous radiologic response patterns following treatment, including pseudo-progression, which was associated with improved tumor control prior to relapse. CONCLUSIONS Our findings point to a key hierarchical role for PD-1 signaling and adaptive immune resistance in the ovarian TME in determining the functional fate of tumor-specific CD8+ T cells, even in the context of robust therapy mediated anti-tumor immunity, as well as the ability of multiple unique patterns of therapeutic response to result in durable tumor control.
Collapse
Affiliation(s)
- A J Robert McGray
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Ruea-Yea Huang
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Cheryl Eppolito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Anthony Miliotto
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Kyle B Stephenson
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Turnstone Biologics, Ottawa, ON, Canada
| | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, NZ, New Zealand
| | - Brian D Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Turnstone Biologics, Ottawa, ON, Canada
| | - Mukund Seshadri
- Department of Dentistry and Maxillofacial Prosthetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Danuta Kozbor
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| |
Collapse
|
19
|
Ranganath N, Sandstrom TS, Burke Schinkel SC, Côté SC, Angel JB. The Oncolytic Virus MG1 Targets and Eliminates Cells Latently Infected With HIV-1: Implications for an HIV Cure. J Infect Dis 2019; 217:721-730. [PMID: 29228368 PMCID: PMC5853232 DOI: 10.1093/infdis/jix639] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 01/09/2023] Open
Abstract
Cells latently infected with human immunodeficiency virus (HIV) evade immune- and drug-mediated clearance. These cells harbor intracellular signaling defects, including impairment of the antiviral type I interferon response. Such defects have also been observed in several cancers and have been exploited for the development of therapeutic oncolytic viruses, including the recombinant Maraba virus (MG1). We therefore hypothesized that MG1 would infect and eliminate cells latently infected with HIV-1, while sparing healthy uninfected cells. Preferential infection and elimination by MG1 was first demonstrated in cell lines latently infected with HIV-1. Following this, a reduction in HIV-1 DNA and inducible HIV-1 replication was observed following MG1 infection of latently infected, resting CD4+ T cells generated using an in vitro model of latency. Last, MG1 infection resulted in a reduction in HIV-1 DNA and inducible HIV-1 replication in memory CD4+ T cells isolated from effectively treated, HIV-1–infected individuals. Our results therefore highlight a novel approach to eliminate the latent HIV-1 reservoir.
Collapse
Affiliation(s)
- Nischal Ranganath
- Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Teslin S Sandstrom
- Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | | | | | - Jonathan B Angel
- Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| |
Collapse
|
20
|
Abstract
The past decades have witnessed significant efforts toward the development of three-dimensional (3D) cell cultures as systems that better mimic in vivo physiology. Today, 3D cell cultures are emerging, not only as a new tool in early drug discovery but also as potential therapeutics to treat disease. In this review, we assess leading 3D cell culture technologies and their impact on drug discovery, including spheroids, organoids, scaffolds, hydrogels, organs-on-chips, and 3D bioprinting. We also discuss the implementation of these technologies in compound identification, screening, and development, ranging from disease modeling to assessment of efficacy and safety profiles.
Collapse
Affiliation(s)
- Ye Fang
- 1 Biochemical Technologies, Corning Research and Development Corporation, Corning Incorporated, Corning, NY, USA
| | - Richard M Eglen
- 2 Corning Life Sciences, Corning Incorporated, Tewksbury, MA, USA
| |
Collapse
|
21
|
Fitzgerald SJ, Janorkar AV, Barnes A, Maranon RO. A new approach to study the sex differences in adipose tissue. J Biomed Sci 2018; 25:89. [PMID: 30509250 PMCID: PMC6278144 DOI: 10.1186/s12929-018-0488-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/15/2018] [Indexed: 11/10/2022] Open
Abstract
Obesity is one of the most invaliding and preventable diseases in the United States. Growing evidence suggests that there are sex differences in obesity in human and experimental animals. However, the specific mechanisms of this disease are unknown. Consequently, there is any particular treatment according to the sex/gender at this time. During the last decade, we observe a rise in the study of adipocyte and the possible mechanisms involved in the different roles of the fat. Furthermore, the effect of sex steroids on the adipocyte is one of the fields that need elucidation. Supporting evidence suggests that sex steroids play an essential role not only in the fat distribution, but also, in its metabolism, proliferation, and function. Thus, using in vitro and in vivo studies will contribute to our fight against this critical health public problem encompassing both sexes. In the present review, we discuss some of the recent advances in the adipocytes and the effect of the sex steroids on the adipose tissue. Also, we propose a new alternative to study the role of sex steroids on adipocyte biology through human adipose-derived stem cells.
Collapse
Affiliation(s)
- Sarah Jayne Fitzgerald
- Department of Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, Jackson, USA
| | - Amol Vijay Janorkar
- Department of Biomedical Materials Science, School of Dentistry, University of Mississippi Medical Center, Jackson, USA
| | - Allison Barnes
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Rodrigo Oscar Maranon
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Medicine/Nephrology, School of Medicine, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Cardio Renal Research Center, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,The Women's Health Research Center, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
22
|
Pol JG, Atherton MJ, Bridle BW, Stephenson KB, Le Boeuf F, Hummel JL, Martin CG, Pomoransky J, Breitbach CJ, Diallo JS, Stojdl DF, Bell JC, Wan Y, Lichty BD. Development and applications of oncolytic Maraba virus vaccines. Oncolytic Virother 2018; 7:117-128. [PMID: 30538968 PMCID: PMC6263248 DOI: 10.2147/ov.s154494] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oncolytic activity of the MG1 strain of the Maraba vesiculovirus has proven efficacy in numerous preclinical cancer models, and relied not only on a direct cytotoxicity but also on the induction of both innate and adaptive antitumor immunity. To further expand tumor-specific T-cell effector and long-lasting memory compartments, we introduced the MG1 virus in a prime-boost cancer vaccine strategy. To this aim, a replication-incompetent adenoviral [Ad] vector together with the oncolytic MG1 have each been armed with a transgene expressing a same tumor antigen. Immune priming with the Ad vaccine subsequently boosted with the MG1 vaccine mounted tumor-specific responses of remarkable magnitude, which significantly prolonged survival in various murine cancer models. Based on these promising results, we validated the safety profile of the Ad:MG1 oncolytic vaccination strategy in nonhuman primates and initiated clinical investigations in cancer patients. Two clinical trials are currently under way (NCT02285816; NCT02879760). The present review will recapitulate the discoveries that led to the development of MG1 oncolytic vaccines from bench to bedside.
Collapse
Affiliation(s)
- Jonathan G Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM), U1138, Paris, France
- Team 11 labelled Ligue Nationale contre le Cancer, Cordeliers Research Center, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Sorbonne Universités/Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Matthew J Atherton
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Fabrice Le Boeuf
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jeff L Hummel
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
- Clinical Trial Division, CANSWERS, Georgetown, ON, Canada
| | | | | | | | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - David F Stojdl
- Turnstone Biologics, Ottawa, ON, Canada,
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - John C Bell
- Turnstone Biologics, Ottawa, ON, Canada,
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
| | - Brian D Lichty
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
- Turnstone Biologics, Ottawa, ON, Canada,
| |
Collapse
|
23
|
Pol JG, Acuna SA, Yadollahi B, Tang N, Stephenson KB, Atherton MJ, Hanwell D, El-Warrak A, Goldstein A, Moloo B, Turner PV, Lopez R, LaFrance S, Evelegh C, Denisova G, Parsons R, Millar J, Stoll G, Martin CG, Pomoransky J, Breitbach CJ, Bramson JL, Bell JC, Wan Y, Stojdl DF, Lichty BD, McCart JA. Preclinical evaluation of a MAGE-A3 vaccination utilizing the oncolytic Maraba virus currently in first-in-human trials. Oncoimmunology 2018; 8:e1512329. [PMID: 30546947 PMCID: PMC6287790 DOI: 10.1080/2162402x.2018.1512329] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple immunotherapeutics have been approved for cancer patients, however advanced solid tumors are frequently refractory to treatment. We evaluated the safety and immunogenicity of a vaccination approach with multimodal oncolytic potential in non-human primates (NHP) (Macaca fascicularis). Primates received a replication-deficient adenoviral prime, boosted by the oncolytic Maraba MG1 rhabdovirus. Both vectors expressed the human MAGE-A3. No severe adverse events were observed. Boosting with MG1-MAGEA3 induced an expansion of hMAGE-A3-specific CD4+ and CD8+ T-cells with the latter peaking at remarkable levels and persisting for several months. T-cells reacting against epitopes fully conserved between simian and human MAGE-A3 were identified. Humoral immunity was demonstrated by the detection of circulating MAGE-A3 antibodies. These preclinical data establish the capacity for the Ad:MG1 vaccination to engage multiple effector immune cell populations without causing significant toxicity in outbred NHPs. Clinical investigations utilizing this program for the treatment of MAGE-A3-positive solid malignancies are underway (NCT02285816, NCT02879760).
Collapse
Affiliation(s)
- Jonathan G Pol
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Sergio A Acuna
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Beta Yadollahi
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Nan Tang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Matthew J Atherton
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - David Hanwell
- Animal Resources Centre, University Health Network, Toronto, ON, Canada
| | | | - Alyssa Goldstein
- Animal Resources Centre, University Health Network, Toronto, ON, Canada
| | - Badru Moloo
- Animal Resources Centre, University Health Network, Toronto, ON, Canada
| | - Patricia V Turner
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Roberto Lopez
- Animal Resources Centre, University Health Network, Toronto, ON, Canada
| | - Sandra LaFrance
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Carole Evelegh
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Galina Denisova
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Robin Parsons
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Jamie Millar
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Gautier Stoll
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Sorbonne Universités/Université Pierre et Marie Curie, Paris, France
| | | | | | | | - Jonathan L Bramson
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - John C Bell
- Turnstone Biologics, Ottawa, ON, Canada.,Ottawa Health Research Institute, Ottawa, ON, Canada
| | - Yonghong Wan
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - David F Stojdl
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Turnstone Biologics, Ottawa, ON, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Turnstone Biologics, Ottawa, ON, Canada
| | - J Andrea McCart
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.,Department of Surgery, Mount Sinai Hospital and University of Toronto, Toronto, Canada
| |
Collapse
|
24
|
Kloker LD, Yurttas C, Lauer UM. Three-dimensional tumor cell cultures employed in virotherapy research. Oncolytic Virother 2018; 7:79-93. [PMID: 30234074 PMCID: PMC6130269 DOI: 10.2147/ov.s165479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Oncolytic virotherapy constitutes an upcoming alternative treatment option for a broad spectrum of cancer entities. However, despite great research efforts, there is still only a single US Food and Drug Administration/European Medicines Agency-approved oncolytic virus available for clinical use. One reason for that is the gap between promising preclinical data and limited clinical success. Since oncolytic viruses are biological agents, they might require more realistic in vitro tumor models than common monolayer tumor cell cultures to provide meaningful predictive preclinical evaluation results. For more realistic invitro tumor models, three-dimensional tumor cell-culture systems can be employed in preclinical virotherapy research. This review provides an overview of spheroid and hydrogel tumor cell cultures, organotypic tumor-tissue slices, organotypic raft cultures, and tumor organoids utilized in the context of oncolytic virotherapy. Furthermore, we also discuss advantages, disadvantages, techniques, and difficulties of these three-dimensional tumor cell-culture systems when applied specifically in virotherapy research.
Collapse
Affiliation(s)
- Linus D Kloker
- Department of Clinical Tumor Biology, University Hospital, University of Tübingen, Tübingen, Germany,
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital, University of Tübingen, Tübingen, Germany
| | - Ulrich M Lauer
- Department of Clinical Tumor Biology, University Hospital, University of Tübingen, Tübingen, Germany, .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Tübingen, Germany,
| |
Collapse
|
25
|
Ma J, Salamoun J, Wipf P, Edwards R, Van Houten B, Qian W. Combination of a thioxodihydroquinazolinone with cisplatin eliminates ovarian cancer stem cell-like cells (CSC-LCs) and shows preclinical potential. Oncotarget 2017; 9:6042-6054. [PMID: 29464053 PMCID: PMC5814193 DOI: 10.18632/oncotarget.23679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/08/2017] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cell-like cells (CSC-LCs) contribute to drug resistance and recurrence of ovarian cancer. Strategies that can eradicate CSC-LCs are expected to substantially improve the outcome of ovarian cancer treatment. We have previously identified a class of thioxodihydroquinazolinone small molecules, which have strong synergistic antitumor activity with platinum drugs, the standard chemotherapeutic agents for ovarian cancer treatment. In the current study, using the activity of aldehyde dehydrogenase (ALDH) as a marker of CSC-LCs, we demonstrated that the combination of thioxodihydroquinazolinone compound 19 with cisplatin is able to diminish ALDH-high CSC-LC populations in both platinum-resistant ovarian cancer cell lines and primary ovarian cancer cells from metastatic ascites of a cisplatin-resistant patient. Compound 19 enhanced the accumulation of intracellular cisplatin in ALDH-high ovarian CSC-LCs. The combination of compound 19 with cisplatin was also able to reduce the sphere-forming capability of cisplatin-resistant ovarian cancer cells. Using a spheroid-based in vitro metastasis model of ovarian cancer, we demonstrated that the co-administration of compound 19 with cisplatin prevents ovarian cancer spheroid cells from attaching to substratum and spreading. In a cisplatin-resistant in vivo intraperitoneal xenograft mouse model, the combination of compound 19 with cisplatin significantly reduced tumor burden, as compared to cisplatin alone. Taken together, our study demonstrated that thioxodihydroquinazolinones represent a new class of agents that in combination with cisplatin are capable of eliminating CSC-LCs in ovarian cancer. Further development of thioxodihydroquinazolinone small molecules may yield a more effective treatment for cisplatin-resistant metastatic ovarian cancer.
Collapse
Affiliation(s)
- Jing Ma
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College of HuaZhong University of Science and Technology, Wuhan 430030, China
| | - Joseph Salamoun
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA.,Accelerated Chemical Discovery Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Robert Edwards
- Department of Obstetrics and Gynecology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Bennett Van Houten
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Wei Qian
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Felt SA, Grdzelishvili VZ. Recent advances in vesicular stomatitis virus-based oncolytic virotherapy: a 5-year update. J Gen Virol 2017; 98:2895-2911. [PMID: 29143726 DOI: 10.1099/jgv.0.000980] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncolytic virus (OV) therapy is an anti-cancer approach that uses viruses that preferentially infect, replicate in and kill cancer cells. Vesicular stomatitis virus (VSV, a rhabdovirus) is an OV that is currently being tested in the USA in several phase I clinical trials against different malignancies. Several factors make VSV a promising OV: lack of pre-existing human immunity against VSV, a small and easy to manipulate genome, cytoplasmic replication without risk of host cell transformation, independence of cell cycle and rapid growth to high titres in a broad range of cell lines facilitating large-scale virus production. While significant advances have been made in VSV-based OV therapy, room for improvement remains. Here we review recent studies (published in the last 5 years) that address 'old' and 'new' challenges of VSV-based OV therapy. These studies focused on improving VSV safety, oncoselectivity and oncotoxicity; breaking resistance of some cancers to VSV; preventing premature clearance of VSV; and stimulating tumour-specific immunity. Many of these approaches were based on combining VSV with other therapeutics. This review also discusses another rhabdovirus closely related to VSV, Maraba virus, which is currently being tested in Canada in phase I/II clinical trials.
Collapse
Affiliation(s)
- Sébastien A Felt
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Valery Z Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
27
|
Olejniczak A, Szaryńska M, Kmieć Z. In vitro characterization of spheres derived from colorectal cancer cell lines. Int J Oncol 2017; 52:599-612. [PMID: 29207035 DOI: 10.3892/ijo.2017.4206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/08/2017] [Indexed: 11/06/2022] Open
Abstract
Spherical cultures (SCs) can be regarded in cancer research as a link between in vitro investigations on cancer lines and in vivo studies of tumor development. SCs are believed to mimic tumor architecture and to be enriched in cancer stem cell-like cells (CSC-like cells). In the present study we characterized colonospheres derived from colorectal cancer (CRC) cell lines, and we confirmed the ability of HCT116 and HT29 cell lines to form spheres within serum-free medium, however, the detailed analysis presented the major differences concerning their characteristics including morphology, phenotype, proliferative potential, distribution in the cell cycle phases and spherogenicity. Moreover, after we magnetically separated CD133+ and CD133- cells we could conduct the analogical analysis as we performed for the original cells. We observed that all cellular fractions unveiled sphere formation capacity, even when cultured in limited number of cells per well and only SCs originated from CD133+ fraction resembled morphologically the parental spheres. Both CD133+ and CD133- subsets derived from HCT116 line were more enriched in cells in G0/G1 phase of the cell cycle in comparison to their HT29 analogues. Additionally, proliferative potential also varied amongst all studied fractions. Surprisingly, 3-D invasion assay revealed that only HCT116-derived populations were able to migrate into extended regions of Matrigel Matrix confirming their higher aggressiveness. Our results provided comprehensive characterization of CRC cell lines culture in adherent and spherical forms and, what seems to be the most advantageous, the comparison of two distinct fractions after magnetic separation. As we found the specific features of cells presented line- and expansion mode-dependency, thus, such complete description might appear crucial before CRC lines would be involved into sophisticated assays, especially focused on potentially novel therapeutic agents targeting CSC-like cells.
Collapse
Affiliation(s)
- Agata Olejniczak
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Magdalena Szaryńska
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
28
|
Tong JG, Valdes YR, Sivapragasam M, Barrett JW, Bell JC, Stojdl D, DiMattia GE, Shepherd TG. Spatial and temporal epithelial ovarian cancer cell heterogeneity impacts Maraba virus oncolytic potential. BMC Cancer 2017; 17:594. [PMID: 28854921 PMCID: PMC5577660 DOI: 10.1186/s12885-017-3600-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 08/23/2017] [Indexed: 12/05/2022] Open
Abstract
Background Epithelial ovarian cancer exhibits extensive interpatient and intratumoral heterogeneity, which can hinder successful treatment strategies. Herein, we investigated the efficacy of an emerging oncolytic, Maraba virus (MRBV), in an in vitro model of ovarian tumour heterogeneity. Methods Four ovarian high-grade serous cancer (HGSC) cell lines were isolated and established from a single patient at four points during disease progression. Limiting-dilution subcloning generated seven additional subclone lines to assess intratumoral heterogeneity. MRBV entry and oncolytic efficacy were assessed among all 11 cell lines. Low-density receptor (LDLR) expression, conditioned media treatments and co-cultures were performed to determine factors impacting MRBV oncolysis. Results Temporal and intratumoral heterogeneity identified two subpopulations of cells: one that was highly sensitive to MRBV, and another set which exhibited 1000-fold reduced susceptibility to MRBV-mediated oncolysis. We explored both intracellular and extracellular mechanisms influencing sensitivity to MRBV and identified that LDLR can partially mediate MRBV infection. LDLR expression, however, was not the singular determinant of sensitivity to MRBV among the HGSC cell lines and subclones. We verified that there were no apparent extracellular factors, such as type I interferon responses, contributing to MRBV resistance. However, direct cell-cell contact by co-culture of MRBV-resistant subclones with sensitive cells restored virus infection and oncolytic killing of mixed population. Conclusions Our data is the first to demonstrate differential efficacy of an oncolytic virus in the context of both spatial and temporal heterogeneity of HGSC cells and to evaluate whether it will constitute a barrier to effective viral oncolytic therapy. Electronic supplementary material The online version of this article (10.1186/s12885-017-3600-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jessica G Tong
- Translational Ovarian Cancer Research Program, London, ON, Canada.,Department of Anatomy & Cell Biology, Western University, London, ON, Canada
| | | | - Milani Sivapragasam
- Translational Ovarian Cancer Research Program, London, ON, Canada.,Department of Anatomy & Cell Biology, Western University, London, ON, Canada
| | - John W Barrett
- Translational Head and Neck Cancer Research Program, London, ON, Canada
| | - John C Bell
- Department of Medicine & Biochemistry, University of Ottawa, Ottawa, ON, Canada.,Department of Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - David Stojdl
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Gabriel E DiMattia
- Translational Ovarian Cancer Research Program, London, ON, Canada.,Department of Biochemistry, Western University, London, ON, Canada.,Department of Oncology, Western University, London, ON, Canada
| | - Trevor G Shepherd
- Translational Ovarian Cancer Research Program, London, ON, Canada. .,Department of Anatomy & Cell Biology, Western University, London, ON, Canada. .,Department of Oncology, Western University, London, ON, Canada. .,Department of Obstetrics & Gynaecology, Western University, London, ON, Canada. .,London Regional Cancer Program, 790 Commissioners Road East, Room A4-836, London, ON, N6A 4L6, Canada.
| |
Collapse
|
29
|
Nounamo B, Liem J, Cannon M, Liu J. Myxoma Virus Optimizes Cisplatin for the Treatment of Ovarian Cancer In Vitro and in a Syngeneic Murine Dissemination Model. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:90-99. [PMID: 28875159 PMCID: PMC5573804 DOI: 10.1016/j.omto.2017.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/06/2017] [Indexed: 12/15/2022]
Abstract
A therapeutic approach to improve treatment outcome of ovarian cancer (OC) in patients is urgently needed. Myxoma virus (MYXV) is a candidate oncolytic virus that infects to eliminate OC cells. We found that in vitro MYXV treatment enhances cisplatin or gemcitabine treatment by allowing lower doses than the corresponding IC50 calculated for primary OC cells. MYXV also affected OC patient ascites-associated CD14+ myeloid cells, one of the most abundant immunological components of the OC tumor environment; without causing cell death, MYXV infection reduces the ability of these cells to secrete cytokines such as IL-10 that are signatures of the immunosuppressive tumor environment. We found that pretreatment with replication-competent but not replication-defective MYXV-sensitized tumor cells to later cisplatin treatments to drastically improve survival in a murine syngeneic OC dissemination model. We thus conclude that infection with replication-competent MYXV before cisplatin treatment markedly enhances the therapeutic benefit of chemotherapy. Treatment with replication-competent MYXV followed by cisplatin potentiated splenocyte activation and IFNγ expression, possibly by T cells, when splenocytes from treated mice were stimulated with tumor cell antigen ex vivo. The impact on immune responses in the tumor environment may thus contribute to the enhanced antitumor activity of combinatorial MYXV-cisplatin treatment.
Collapse
Affiliation(s)
- Bernice Nounamo
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA
| | - Jason Liem
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA
| | - Martin Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA.,The Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
30
|
Szarynska M, Olejniczak A, Wierzbicki P, Kobiela J, Laski D, Sledzinski Z, Adrych K, Guzek M, Kmiec Z. FasR and FasL in colorectal cancer. Int J Oncol 2017; 51:975-986. [PMID: 28766682 DOI: 10.3892/ijo.2017.4083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/11/2017] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common solid organ cancers prevalent worldwide causing, in spite of advancing therapeutic methodology, high rate of patient mortality, especially due to metastasis development. The cancer stem cell (CSC) theory of tumor growth indicates that CSCs within the tumor mass have great capacity to initiate and sustain tumor growth. Following the suggestion that Fas signaling can be engaged in apoptosis, tumor maintenance, senescence or DICE (death induced by CD95 or CD95L elimination), the attempts to broaden the knowledge concerning the relationships between CSCs features and FasR/FasL appeared to be necessary. The most important advantage of our study was the simultaneously analysis of CSCs from commonly used CRC lines (HCT116 and HT29) and tumor fragments collected from CRC patients. Moreover, the sphere-promoting expansion of CRC lines brought a specific three-dimensional specific environment for CSC exploration. We further investigated the function of Fas signaling in CRC lines depending on the culture mode as we incubated HCT116 and HT29 cells with anti-FasR agonistic antibodies. It appeared to act in a line-dependent and culture mode-dependent manner and influenced some particular features of CSCs such as spherogenicity, proliferation and phenotype. Additionally, the analysis of mRNA level showed that disease progression is associated with significantly increased expression of FasR and/or FasL. In conclusion, our observation seems to confirm that spherical model of cancer lines is more reliable for some sophisticated analysis because of their greater resemblance to the CSCs from human CRC samples in comparison to commonly used adherent cells, at least according to aspects of their biology analyzed in this study. That can be extended to the resemblance of in vitro sphere forming conditions to the in vivo environment. However, the greatest difference concerns the level of apoptosis, thus, this issue require further experiments.
Collapse
Affiliation(s)
- Magdalena Szarynska
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Agata Olejniczak
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Piotr Wierzbicki
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Jaroslaw Kobiela
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Invasive Medicine Centre, 80-214 Gdansk, Poland
| | - Dariusz Laski
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Invasive Medicine Centre, 80-214 Gdansk, Poland
| | - Zbigniew Sledzinski
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Invasive Medicine Centre, 80-214 Gdansk, Poland
| | - Krystian Adrych
- Department of Hepatology and Gastroenterology, Medical University of Gdansk, Invasive Medicine Centre, 80-214 Gdansk, Poland
| | - Marek Guzek
- Department of Hepatology and Gastroenterology, Medical University of Gdansk, Invasive Medicine Centre, 80-214 Gdansk, Poland
| | - Zbigniew Kmiec
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
31
|
Orzechowska BU, Jędryka M, Zwolińska K, Matkowski R. VSV based virotherapy in ovarian cancer: the past, the present and …future? J Cancer 2017; 8:2369-2383. [PMID: 28819441 PMCID: PMC5560156 DOI: 10.7150/jca.19473] [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: 02/03/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
The standard approach to treating patients with advanced epithelial ovarian cancer (EOC) after primary debulking surgery remains taxane and platinum-based chemotherapy. Despite treatment with this strategy, the vast majority of patients relapse and develop drug-resistant metastatic disease that may be driven by cancer stem cells (CSCs) or cancer initiating cells (CICs). Oncolytic viruses circumvent typical drug-resistance mechanisms, therefore they may provide a safe and effective alternative treatment for chemotherapy-resistant CSCs/CICs. Among oncolytic viruses vesicular stomatitis virus (VSV) has demonstrated oncolysis and preferential replication in cancer cells. In this review, we summarize the recent findings regarding existing knowledge on biology of the ovarian cancer and the role of ovarian CSCs (OCSCs) in tumor dissemination and chemoresistance. In addition we also present an overview of recent advances in ovarian cancer therapies with oncolytic viruses (OV). We focus particularly on key genetic or immune response pathways involved in tumorigenesis in ovarian cancer which facilitate oncolytic activity of vesicular stomatitis virus (VSV). We highlight the prospects of targeting OCSCs with VSV. The importance of testing an emerging ovarian cancer animal models and ovarian cancer cell culture conditions influencing oncolytic efficacy of VSV is also addressed.
Collapse
Affiliation(s)
- Beata Urszula Orzechowska
- Laboratory of Virology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Marcin Jędryka
- Division of Surgical Oncology, Gynaecological Oncology, Chemotherapy and Department of Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wrocław, Poland
- Lower Silesian Oncology Centre, Wroclaw, Plac Hirszfelda 12, 53-413 Wrocław, Poland
| | - Katarzyna Zwolińska
- Laboratory of Virology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wroclaw, Poland
| | - Rafał Matkowski
- Division of Surgical Oncology, Gynaecological Oncology, Chemotherapy and Department of Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53-413 Wrocław, Poland
- Lower Silesian Oncology Centre, Wroclaw, Plac Hirszfelda 12, 53-413 Wrocław, Poland
| |
Collapse
|
32
|
Fang Y, Eglen RM. Three-Dimensional Cell Cultures in Drug Discovery and Development. SLAS DISCOVERY 2017. [DOI: 10.1177/2472555217696795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ye Fang
- Biochemical Technologies, Corning Research and Development Corporation, Corning Incorporated, Corning, NY, USA
| | | |
Collapse
|
33
|
Alkayyal AA, Tai LH, Kennedy MA, de Souza CT, Zhang J, Lefebvre C, Sahi S, Ananth AA, Mahmoud AB, Makrigiannis AP, Cron GO, Macdonald B, Marginean EC, Stojdl DF, Bell JC, Auer RC. NK-Cell Recruitment Is Necessary for Eradication of Peritoneal Carcinomatosis with an IL12-Expressing Maraba Virus Cellular Vaccine. Cancer Immunol Res 2017; 5:211-221. [PMID: 28159747 DOI: 10.1158/2326-6066.cir-16-0162] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/09/2016] [Accepted: 01/11/2017] [Indexed: 11/16/2022]
Abstract
Despite improvements in chemotherapy and radical surgical debulking, peritoneal carcinomatosis (PC) remains among the most common causes of death from abdominal cancers. Immunotherapies have been effective for selected solid malignancies, but their potential in PC has been little explored. Here, we report that intraperitoneal injection of an infected cell vaccine (ICV), consisting of autologous tumor cells infected ex vivo with an oncolytic Maraba MG1 virus expressing IL12, promotes the migration of activated natural killer (NK) cells to the peritoneal cavity in response to the secretion of IFNγ-induced protein-10 (IP-10) from dendritic cells. The recruitment of cytotoxic, IFNγ-secreting NK cells was associated with reduced tumor burden and improved survival in a colon cancer model of PC. Even in mice with bulky PC (tumors > 8 mm), a complete radiologic response was demonstrated within 8 to14 weeks, associated with 100% long-term survival. The impact of MG1-IL12-ICV upon NK-cell recruitment and function observed in the murine system was recapitulated in human lymphocytes exposed to human tumor cell lines infected with MG1-IL12. These findings suggest that an MG1-IL12-ICV is a promising therapy that could provide benefit to the thousands of patients diagnosed with PC each year. Cancer Immunol Res; 5(3); 211-21. ©2017 AACR.
Collapse
Affiliation(s)
- Almohanad A Alkayyal
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute.,Department of Laboratory Medicine, University of Tabuk, Tabuk, Saudi Arabia.,Department of BMI, University of Ottawa, Ottawa, Ontario, Canada
| | - Lee-Hwa Tai
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute
| | - Michael A Kennedy
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute
| | | | - Jiqing Zhang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute
| | - Charles Lefebvre
- Apoptosis Research Centre, CHEO Research Institute, Ottawa, Ontario, Canada
| | - Shalini Sahi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute
| | - Abhirami A Ananth
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute.,Department of BMI, University of Ottawa, Ottawa, Ontario, Canada
| | - Ahmad Bakur Mahmoud
- Department of BMI, University of Ottawa, Ottawa, Ontario, Canada.,Department of Medical Technology, Taibah University, Medina, Saudi Arabia
| | | | - Greg O Cron
- Department of Medical Imaging, University of Ottawa, Ottawa, Ontario, Canada.,Department of Radiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Blair Macdonald
- Department of Medical Imaging, University of Ottawa, Ottawa, Ontario, Canada
| | - E Celia Marginean
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David F Stojdl
- Department of BMI, University of Ottawa, Ottawa, Ontario, Canada.,Apoptosis Research Centre, CHEO Research Institute, Ottawa, Ontario, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute.,Department of BMI, University of Ottawa, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute. .,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|