1
|
Zinovieva M, Ryapolova A, Karabelsky A, Minskaia E. Oncolytic Vesicular Stomatitis Virus: Optimisation Strategies for Anti-Cancer Therapies. FRONT BIOSCI-LANDMRK 2024; 29:374. [PMID: 39614430 DOI: 10.31083/j.fbl2911374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024]
Abstract
Oncolytic viruses (OVs) represent a targeted anti-cancer therapy approach due to their ability not only to selectively infect and destroy malignant cells but also to induce an immune response. Vesicular stomatitis virus (VSV) offers a promising platform due to its low prevalence and pathogenicity in humans, lack of pre-existing immunity, easily manipulated genome, rapid growth to high titers in a broad range of cell lines, and inability to integrate into the host genome. However, despite its many advantages, many unresolved problems remain: problematic production based on the reverse genetics system, oncological selectivity, and the overall effectiveness of VSV monotherapy. This review will discuss various attempts at viral genome modifications aimed at improving the oncolytic properties of VSV. These strategies include inhibition of viral genes, modification of genes responsible for targeting cancer cells over healthy ones, insertion of foreign genes for boosting immune response, and changing the order of viral and inserted foreign genes. In addition, possible ways to improve VSV-based anti-tumor therapy and achieve higher efficiency will be considered by evaluating the effectiveness of various delivery methods as well as discussing treatment options by combining VSV with other groups of anticancer drugs.
Collapse
Affiliation(s)
- Margarita Zinovieva
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia Ryapolova
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Alexander Karabelsky
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ekaterina Minskaia
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
2
|
Xu L, Sun H, Lemoine NR, Xuan Y, Wang P. Oncolytic vaccinia virus and cancer immunotherapy. Front Immunol 2024; 14:1324744. [PMID: 38283361 PMCID: PMC10811104 DOI: 10.3389/fimmu.2023.1324744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Oncolytic virotherapy (OVT) is a promising form of cancer treatment that uses genetically engineered viruses to replicate within cancer cells and trigger anti-tumor immune response. In addition to killing cancer cells, oncolytic viruses can also remodel the tumor microenvironment and stimulate a long-term anti-tumor immune response. Despite achieving positive results in cellular and organismal studies, there are currently only a few approved oncolytic viruses for clinical use. Vaccinia virus (VACV) has emerged as a potential candidate due to its ability to infect a wide range of cancer cells. This review discusses the mechanisms, benefits, and clinical trials of oncolytic VACVs. The safety and efficacy of different viral backbones are explored, as well as the effects of oncolytic VACVs on the tumor microenvironment. The potential combination of oncolytic VACVs with immunotherapy or traditional therapies is also highlighted. The review concludes by addressing prospects and challenges in the field of oncolytic VACVs, with the aim of promoting further research and application in cancer therapy.
Collapse
Affiliation(s)
- Lihua Xu
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huihui Sun
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Nicholas R. Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Yujing Xuan
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Hamdan F, Fusciello M, Cerullo V. Personalizing Oncolytic Virotherapy. Hum Gene Ther 2023; 34:870-877. [PMID: 37698876 DOI: 10.1089/hum.2023.122] [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] [Indexed: 09/13/2023] Open
Abstract
The use of oncolytic viruses has become an attractive tool in the clinics for the treatment of various tumor types. Such viruses are genetically modified to conditionally replicate in malignant cells while unharming healthy cells. This platform offers a highly specific tumor killing with exceptional safety profiles. However, the use of oncolytic viruses as sole oncolytic platforms has not achieved full tumor clearance in murine models and in the clinics. In fact, the formation of anti-tumor immune responses is attributed to the effectiveness of oncolytic viruses. In this review, we will discuss the various strategies that scientists have employed to enhance the anti-tumor immune responses driven by oncolytic viruses. Moreover, focus will be drawn into personalizing such anti-tumor responses by the addition of tumor-associated peptides.
Collapse
Affiliation(s)
- Firas Hamdan
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
- Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Manlio Fusciello
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
- Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
- Drug Delivery, Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Feola S, Russo S, Ylösmäki E, Cerullo V. Oncolytic ImmunoViroTherapy: A long history of crosstalk between viruses and immune system for cancer treatment. Pharmacol Ther 2021; 236:108103. [PMID: 34954301 DOI: 10.1016/j.pharmthera.2021.108103] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
Cancer Immunotherapy relies on harnessing a patient's immune system to fine-tune specific anti-tumor responses and ultimately eradicate cancer. Among diverse therapeutic approaches, oncolytic viruses (OVs) have emerged as a novel form of cancer immunotherapy. OVs are a naturally occurring or genetically modified class of viruses able to selectively kill cancer cells, leaving healthy cells unharmed; in the last two decades, the role of OVs has been redefined to act beyond their oncolytic activity. Indeed, the immunogenic cancer cell death mediated by OVs induces the release of tumor antigens that in turn induces anti-tumor immunity, allowing OVs to act as in situ therapeutic cancer vaccines. Additionally, OVs can be engineered for intratumoral delivery of immunostimulatory molecules such as tumor antigens or cytokines to further enhance anti-tumor response. Moreover, OVs can be used in combination with other cancer immunotherapeutic approaches such as Immune Checkpoint Inhibitors and CAR-T cells. The current review first defines the three main mechanisms of action (MOA) of OVs currently used in cancer therapy that are: i) Oncolysis, ii) OV-induced cancer-specific immune activation, and iii) Exploiting pre-existing anti-viral immunity to enhance cancer therapy. Secondly, we focus on how OVs can induce and/or improve anti-cancer immunity in a specific or unspecific fashion, highlighting the importance of these approaches. Finally, the last part of the review analyses OVs combined with other cancer immunotherapies, revising present and future clinical applications.
Collapse
Affiliation(s)
- S Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland; TRIMM, Translational Immunology Research Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Finland
| | - S Russo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland; TRIMM, Translational Immunology Research Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Finland
| | - E Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland; TRIMM, Translational Immunology Research Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Finland
| | - V Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland; TRIMM, Translational Immunology Research Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Finland; Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University Federico II, S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
5
|
Boagni DA, Ravirala D, Zhang SX. Current strategies in engaging oncolytic viruses with antitumor immunity. Mol Ther Oncolytics 2021; 22:98-113. [PMID: 34514092 PMCID: PMC8411207 DOI: 10.1016/j.omto.2021.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oncolytic virotherapy has produced promising yet limited results in preclinical and clinical studies. Besides direct oncolytic activity, a significant therapeutic mechanism of oncolytic virotherapy is the induction of tumor-specific immunity. Consequently, the efficacy of oncolytic viruses can be improved by the insertion of immune stimulator genes and rational combinatorial therapy with other immunotherapies. This article reviews recent efforts on arming oncolytic viruses with a variety of immune stimulator molecules, immune cell engagers, and other immune potentiating molecules. We outline what is known about the mechanisms of action and the corresponding results. The review also discusses recent preclinical and clinical studies of combining oncolytic virotherapy with immune-checkpoint inhibitors and the role of oncolytic virotherapy in changing the tumor microenvironment.
Collapse
Affiliation(s)
- Drew Ashton Boagni
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Divya Ravirala
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Shaun Xiaoliu Zhang
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
6
|
Simsekoglu MF, Kaleler I, Onal B, Demirdag C, Citgez S, Uslu E, Erozenci A, Talat Z. Do urinary mast cell mediators predict immune response to BCG in patients with primary high-grade non-muscle invasive bladder cancer? Int J Clin Pract 2021; 75:e13959. [PMID: 33369059 DOI: 10.1111/ijcp.13959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Mast cells play a critical role in cancer-associated immunity. We aimed to determine the predictive value of urinary mast cell mediators in patients with non-muscle invasive bladder cancer (NMIBC) treated with Bacillus Calmette-Guérin (BCG) immunotherapy. METHODS In this prospective study, 19 patients who received immunotherapy because of NMIBC (Group 1) and 19 healthy participants (Group 2) were enrolled. Urine samples were collected to assay N-methylhistamine, histamine, and tryptase levels immediately before the first BCG instillation, immediately after the third and sixth instillations, and 4 weeks after the sixth instillation in Group 1 and at a single visit in Group 2. The changes in urinary markers because of BCC response, BCG instillation, and the presence of NMIBC were assessed. RESULTS The average age was 56.1 ± 10.5 years in Group 1 and 52.6 ± 9.7 years in Group 2. Fourteen patients had high-grade Ta tumours and five had T1 tumours. While 12 patients had responded to the BCG, seven patients did not respond to the BCG. There was no correlation between mast cell mediators and BCG response. The N-methylhistamine and histamine levels significantly increased with the onset of immunotherapy, and N-methylhistamine levels significantly decreased when immunotherapy was terminated (P < .05). The pre-BCG estimated marginal mean values of N-methylhistamine were significantly higher in Group 1 than in Group 2 (P < .05). CONCLUSIONS Our study is the first to identify the changes in mast cell mediators with the onset of immunotherapy and in presence of bladder cancer. However, these mediators cannot predict patients' response to immunotherapy.
Collapse
Affiliation(s)
- Muhammed Fatih Simsekoglu
- Department of Urology, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Islim Kaleler
- Department of Biochemistry, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Bulent Onal
- Department of Urology, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Cetin Demirdag
- Department of Urology, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Sinharib Citgez
- Department of Urology, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Ezel Uslu
- Department of Biochemistry, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Ahmet Erozenci
- Department of Urology, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Zubeyr Talat
- Department of Urology, Istanbul University- Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| |
Collapse
|
7
|
Zafar S, Basnet S, Launonen IM, Quixabeira DCA, Santos J, Hemminki O, Malmstedt M, Cervera-Carrascon V, Aronen P, Kalliokoski R, Havunen R, Rannikko A, Mirtti T, Matikainen M, Kanerva A, Hemminki A. Oncolytic Adenovirus Type 3 Coding for CD40L Facilitates Dendritic Cell Therapy of Prostate Cancer in Humanized Mice and Patient Samples. Hum Gene Ther 2021; 32:192-202. [PMID: 33050725 PMCID: PMC10112462 DOI: 10.1089/hum.2020.222] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dendritic cell (DC)-based vaccines have shown some degree of success for the treatment of prostate cancer (PC). However, the highly immunosuppressive tumor microenvironment leads to DC dysfunction, which has limited the effectiveness of these vaccines. We hypothesized that use of a fully serotype 3 oncolytic adenovirus (Ad3-hTERT-CMV-hCD40L; TILT-234) could stimulate DCs in the prostate tumor microenvironment by expressing CD40L. Activated DCs would then activate cytotoxic T cells against the tumor, resulting in therapeutic immune responses. Oncolytic cell killing due to cancer cell-specific virus replication adds to antitumor effects but also enhances the immunological effect by releasing tumor epitopes for sampling by DC, in the presence of danger signals. In this study, we evaluated the companion effect of Ad3-hTERT-CMV-hCD40L and DC-therapy in a humanized mouse model and PC histocultures. Treatment with Ad3-hTERT-CMV-hCD40L and DC resulted in enhanced antitumor responses in vivo. Treatment of established histocultures with Ad3-hTERT-CMV-hCD40L induced DC maturation and notable increase in proinflammatory cytokines. In conclusion, Ad3-hTERT-CMV-hCD40L is able to modulate an immunosuppressive prostate tumor microenvironment and improve the effectiveness of DC vaccination in PC models and patient histocultures, setting the stage for clinical translation.
Collapse
Affiliation(s)
- Sadia Zafar
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Inga-Maria Launonen
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Dafne Carolina Alves Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Joao Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Otto Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,Division of Urology, Department of Surgery, University Health Network and University of Toronto, Toronto, Canada.,Department of Urology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | | | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Pasi Aronen
- Biostatistics Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Antti Rannikko
- Department of Urology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | | | | | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland.,Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
8
|
Pelin A, Boulton S, Tamming LA, Bell JC, Singaravelu R. Engineering vaccinia virus as an immunotherapeutic battleship to overcome tumor heterogeneity. Expert Opin Biol Ther 2020; 20:1083-1097. [PMID: 32297534 DOI: 10.1080/14712598.2020.1757066] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Immunotherapy is a rapidly evolving area of cancer therapeutics aimed at driving a systemic immune response to fight cancer. Oncolytic viruses (OVs) are at the cutting-edge of innovation in the immunotherapy field. Successful OV platforms must be effective in reshaping the tumor microenvironment and controlling tumor burden, but also be highly specific to avoid off-target side effects. Large DNA viruses, like vaccinia virus (VACV), have a large coding capacity, enabling the encoding of multiple immunostimulatory transgenes to reshape the tumor immune microenvironment. VACV-based OVs have shown promising results in both pre-clinical and clinical studies, including safe and efficient intravenous delivery to metastatic tumors. AREA COVERED This review summarizes attenuation strategies to generate a recombinant VACV with optimal tumor selectivity and immunogenicity. In addition, we discuss immunomodulatory transgenes that have been introduced into VACV and summarize their effectiveness in controlling tumor burden. EXPERT OPINION VACV encodes several immunomodulatory genes which aid the virus in overcoming innate and adaptive immune responses. Strategic deletion of these virulence factors will enable an optimal balance between viral persistence and immunogenicity, robust tumor-specific expression of payloads and promotion of a systemic anti-cancer immune response. Rational selection of therapeutic transgenes will maximize the efficacy of OVs and their synergy in combinatorial immunotherapy schemes.
Collapse
Affiliation(s)
- Adrian Pelin
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Stephen Boulton
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Levi A Tamming
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Ragunath Singaravelu
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| |
Collapse
|
9
|
Pelin A, Foloppe J, Petryk J, Singaravelu R, Hussein M, Gossart F, Jennings VA, Stubbert LJ, Foster M, Storbeck C, Postigo A, Scut E, Laight B, Way M, Erbs P, Le Boeuf F, Bell JC. Deletion of Apoptosis Inhibitor F1L in Vaccinia Virus Increases Safety and Oncolysis for Cancer Therapy. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:246-252. [PMID: 31428674 PMCID: PMC6695278 DOI: 10.1016/j.omto.2019.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022]
Abstract
Vaccinia virus (VACV) possesses a great safety record as a smallpox vaccine and has been intensively used as an oncolytic virus against various types of cancer over the past decade. Different strategies were developed to make VACV safe and selective to cancer cells. Leading clinical candidates, such as Pexa-Vec, are attenuated through deletion of the viral thymidine kinase (TK) gene, which limits virus growth to replicate in cancer tissue. However, tumors are not the only tissues whose metabolic activity can overcome the lack of viral TK. In this study, we sought to further increase the tumor-specific replication and oncolytic potential of Copenhagen strain VACV ΔTK. We show that deletion of the anti-apoptosis viral gene F1L not only increases the safety of the Copenhagen ΔTK virus but also improves its oncolytic activity in an aggressive glioblastoma model. The additional loss of F1L does not affect VACV replication capacity, yet its ability to induce cancer cell death is significantly increased. Our results also indicate that cell death induced by the Copenhagen ΔTK/F1L mutant releases more immunogenic signals, as indicated by increased levels of IL-1β production. A cytotoxicity screen in an NCI-60 panel shows that the ΔTK/F1L virus induces faster tumor cell death in different cancer types. Most importantly, we show that, compared to the TK-deleted virus, the ΔTK/F1L virus is attenuated in human normal cells and causes fewer pox lesions in murine models. Collectively, our findings describe a new oncolytic vaccinia deletion strain that improves safety and increases tumor cell killing.
Collapse
Affiliation(s)
- Adrian Pelin
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Johann Foloppe
- Transgene S.A., 400 Boulevard Gonthier d'Andernach, 67405 Illkirch-Graffenstaden, France
| | - Julia Petryk
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Ragunath Singaravelu
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Marian Hussein
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Florian Gossart
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Victoria A Jennings
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Lawton J Stubbert
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Madison Foster
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Christopher Storbeck
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Antonio Postigo
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, England, UK
| | - Elena Scut
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Brian Laight
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, England, UK
| | - Philippe Erbs
- Transgene S.A., 400 Boulevard Gonthier d'Andernach, 67405 Illkirch-Graffenstaden, France
| | - Fabrice Le Boeuf
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - John C Bell
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
10
|
Governa V, Brittoli A, Mele V, Pinamonti M, Terracciano L, Muenst S, Iezzi G, Spagnoli GC, Zajac P, Trella E. A replication-incompetent CD154/40L recombinant vaccinia virus induces direct and macrophage-mediated antitumor effects in vitro and in vivo. Oncoimmunology 2019; 8:e1568162. [PMID: 31069131 PMCID: PMC6492963 DOI: 10.1080/2162402x.2019.1568162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/30/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
CD40 triggering may result in antitumor effects of potentially high clinical relevance. To gain insights important for patient selection and to identify adequate targeting techniques, we investigated CD40 expression in human cancer tissues and generated a replication-incompetent recombinant vaccinia virus expressing CD40 ligand (rVV40L). Its effects were explored in vitro and in vivo upon direct CD40 targeting on malignant cells or macrophage activation. CD40 expression was analyzed by immunohistochemistry in tumor and stromal cells in a multi-tumor array including 836 specimens from 27 different tumor types. Established tumor cell lines were used to explore the capacity of rVV40L to induce malignant cell apoptosis and modulate functional profiles of polarized macrophages. CD40 expression was detectable in significantly higher numbers of stromal as compared to malignant cells in lung and breast cancers. CD40 ligation following rVV40L infection induced apoptosis in CD40(+) cancer cells, but only in the presence of intact specific signal transduction chain. Importantly, rVV40L infection promoted the induction of TNF-α-dependent antitumor activity of M1-like macrophages directed against CD40(-) targets. CD40-activated M1-like macrophages also displayed enhanced ability to CXCL10-dependently recruit CD8+ T cells and to efficiently present cancer cell intracellular antigens through cross-priming. Moreover, rVV-driven CD40L expression partially “re-educated” M2-like macrophages, as suggested by detectable CXCL10 and IL-12 production. Most importantly, we observed that intra-tumoral injection of rVV40L-infected human macrophages inhibits progression of human CD40(-) tumors in vivo. First evidences of anticancer activity of rVV40L strongly encourage further evaluations.
Collapse
Affiliation(s)
- Valeria Governa
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Alvaro Brittoli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valentina Mele
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Maurizio Pinamonti
- Unit of Pathology, Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Luigi Terracciano
- Institute of Pathology, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Simone Muenst
- Institute of Pathology, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Giandomenica Iezzi
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Department of Surgery, Ente Ospedaliero Cantonale and Università della Svizzera Italiana, Lugano, Switzerland
| | - Giulio Cesare Spagnoli
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.,Consiglio Nazionale delle Ricerche, Institute of Translational Pharmacology, Rome, Italy
| | - Paul Zajac
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Emanuele Trella
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
11
|
Zafar S, Sorsa S, Siurala M, Hemminki O, Havunen R, Cervera-Carrascon V, Santos JM, Wang H, Lieber A, De Gruijl T, Kanerva A, Hemminki A. CD40L coding oncolytic adenovirus allows long-term survival of humanized mice receiving dendritic cell therapy. Oncoimmunology 2018; 7:e1490856. [PMID: 30386680 PMCID: PMC6207416 DOI: 10.1080/2162402x.2018.1490856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are crucial players in promoting immune responses. Logically, adoptive DC therapy is a promising approach in cancer immunotherapy. One of the major obstacles in cancer immunotherapy in general is the immunosuppressive tumor microenvironment, which hampers the maturation and activation of DCs. Therefore, human clinical outcomes with DC therapy alone have been disappointing. In this study, we use fully serotype 3 oncolytic adenovirus Ad3-hTERT-CMV-hCD40L, expressing human CD40L, to modulate the tumor microenvironment with subsequently improved function of DCs. We evaluated the synergistic effects of Ad3-hTERT-CMV-hCD40L and DCs in the presence of human peripheral blood mononuclear cells ex vivo and in vivo. Tumors treated with Ad3-hTERT-CMV-hCD40L and DCs featured greater antitumor effect compared with unarmed virus or either treatment alone. 100% of humanized mice survived to the end of the experiment, while mice in all other groups died by day 88. Moreover, adenovirally-delivered CD40L induced activation of DCs, leading to induction of Th1 immune responses. These results support clinical trials with Ad3-hTERT-CMV-hCD40L in patients receiving DC therapy.
Collapse
Affiliation(s)
- Sadia Zafar
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- Division of Urology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - João Manuel Santos
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Hongjie Wang
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Andre Lieber
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Tanja De Gruijl
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Anna Kanerva
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
12
|
de Graaf JF, de Vor L, Fouchier RAM, van den Hoogen BG. Armed oncolytic viruses: A kick-start for anti-tumor immunity. Cytokine Growth Factor Rev 2018; 41:28-39. [PMID: 29576283 PMCID: PMC7108398 DOI: 10.1016/j.cytogfr.2018.03.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 12/16/2022]
Abstract
Oncolytic viruses (OVs), viruses that specifically result in killing tumor cells, represent a promising class of cancer therapy. Recently, the focus in the OV therapy field has shifted from their direct oncolytic effect to their immune stimulatory effect. OV therapy can function as a "kick start" for the antitumor immune response by releasing tumor associated antigens and release of inflammatory signals. Combining OVs with immune modulators could enhance the efficacy of both immune and OV therapies. Additionally, genetic engineering of OVs allows local expression of immune therapeutics, thereby reducing related toxicities. Different options to modify the tumor microenvironment in combination with OV therapy have been explored. The possibilities and obstacles of these combinations will be discussed in this review.
Collapse
Affiliation(s)
- J F de Graaf
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.
| | - L de Vor
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.
| | - R A M Fouchier
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.
| | | |
Collapse
|
13
|
Pelin A, Wang J, Bell J, Le Boeuf F. The importance of imaging strategies for pre-clinical and clinical in vivo distribution of oncolytic viruses. Oncolytic Virother 2018; 7:25-35. [PMID: 29637059 PMCID: PMC5880516 DOI: 10.2147/ov.s137159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) are an emergent and unique therapy for cancer patients. Similar to chemo- and radiation therapy, OV can lyse (kill) cancer cell directly. In general, the advantages of OVs over other treatments are primarily: a higher safety profile (as shown by less adverse effects), ability to replicate, transgene(s) delivery, and stimulation of a host’s immune system against cancer. The latter has prompted successful use of OVs with other immunotherapeutic strategies in a synergistic manner. In spite of extended testing in pre-clinical and clinical setting, using biologically derived therapeutics like virus always raises potential concerns about safety (replication at non-intended locations) and bio-availability of the product. Recent advent in in vivo imaging techniques dramatically improves the convenience of use, quality of pictures, and amount of information acquired. Easy assessing of safety/localization of the biotherapeutics like OVs became a new potential weapon in the physician’s arsenal to improve treatment outcome. Given that OVs are typically replicating, in vivo imaging can also track virus replication and persistence as well as precisely mapping tumor tissues presence. This review discusses the importance of imaging in vivo in evaluating OV efficacy, as well as currently available tools and techniques.
Collapse
Affiliation(s)
- Adrian Pelin
- Department of Biochemistry, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Ottawa Hospital Research Institute, Center for Cancer Therapeutics, Ottawa, ON, Canada
| | - Jiahu Wang
- Ottawa Hospital Research Institute, Center for Cancer Therapeutics, Ottawa, ON, Canada.,Genvira Biosciences, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - John Bell
- Department of Biochemistry, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Ottawa Hospital Research Institute, Center for Cancer Therapeutics, Ottawa, ON, Canada
| | - Fabrice Le Boeuf
- Ottawa Hospital Research Institute, Center for Cancer Therapeutics, Ottawa, ON, Canada
| |
Collapse
|
14
|
Francis L, Guo ZS, Liu Z, Ravindranathan R, Urban JA, Sathaiah M, Magge D, Kalinski P, Bartlett DL. Modulation of chemokines in the tumor microenvironment enhances oncolytic virotherapy for colorectal cancer. Oncotarget 2017; 7:22174-85. [PMID: 26956047 PMCID: PMC5008353 DOI: 10.18632/oncotarget.7907] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/20/2016] [Indexed: 12/31/2022] Open
Abstract
An oncolytic poxvirus such as vvDD-CXCL11 can generate potent systemic antitumor immunity as well as targeted oncolysis, yet the antitumor effect is limited probably due to limited homing to and suppressed activity of tumor-specific adaptive immune cells in the tumor microenvironment (TME). We reasoned that a chemokine modulating (CKM) drug cocktail, consisting of IFN-α, poly I:C, and a COX-2 inhibitor, may skew the chemokine (CK) and cytokine profile into a favorable one in the TME, and this pharmaceutical modulation would enhance both the trafficking into and function of antitumor immune cells in the TME, thus increasing therapeutic efficacy of the oncolytic virus. In this study we show for the first time in vivo that the CKM modulates the CK microenvironment but it does not modulate antitumor immunity by itself in a MC38 colon cancer model. Sequential treatment with the virus and then CKM results in the upregulation of Th1-attracting CKs and reduction of Treg-attracting CKs (CCL22 and CXCL12), concurrent with enhanced trafficking of tumor-specific CD8+ T cells and NK cells into the TME, thus resulting in the most significant antitumor activity and long term survival of tumor-bearing mice. This novel combined regimen, with the oncolytic virus (vvDD-CXCL11) inducing direct oncolysis and eliciting potent antitumor immunity, and the CKM inducing a favorable chemokine profile in the TME that promotes the trafficking and function of antitumor Tc1/Th1 and NK cells, may have great utility for oncolytic immunotherapy for cancer.
Collapse
Affiliation(s)
- Lily Francis
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zong Sheng Guo
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zuqiang Liu
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roshni Ravindranathan
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julie A Urban
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Magesh Sathaiah
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deepa Magge
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pawel Kalinski
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L Bartlett
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Koval O, Kochneva G, Tkachenko A, Troitskaya O, Sivolobova G, Grazhdantseva A, Nushtaeva A, Kuligina E, Richter V. Recombinant Vaccinia Viruses Coding Transgenes of Apoptosis-Inducing Proteins Enhance Apoptosis But Not Immunogenicity of Infected Tumor Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3620510. [PMID: 28951871 PMCID: PMC5603130 DOI: 10.1155/2017/3620510] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/17/2017] [Accepted: 07/26/2017] [Indexed: 12/16/2022]
Abstract
Genetic modifications of the oncolytic vaccinia virus (VV) improve selective tumor cell infection and death, as well as activation of antitumor immunity. We have engineered a double recombinant VV, coding human GM-CSF, and apoptosis-inducing protein apoptin (VV-GMCSF-Apo) for comparing with the earlier constructed double recombinant VV-GMCSF-Lact, coding another apoptosis-inducing protein, lactaptin, which activated different cell death pathways than apoptin. We showed that both these recombinant VVs more considerably activated a set of critical apoptosis markers in infected cells than the recombinant VV coding GM-CSF alone (VV-GMCSF-dGF): these were phosphatidylserine externalization, caspase-3 and caspase-7 activation, DNA fragmentation, and upregulation of proapoptotic protein BAX. However, only VV-GMCSF-Lact efficiently decreased the mitochondrial membrane potential of infected cancer cells. Investigating immunogenic cell death markers in cancer cells infected with recombinant VVs, we demonstrated that all tested recombinant VVs were efficient in calreticulin and HSP70 externalization, decrease of cellular HMGB1, and ATP secretion. The comparison of antitumor activity against advanced MDA-MB-231 tumor revealed that both recombinants VV-GMCSF-Lact and VV-GMCSF-Apo efficiently delay tumor growth. Our results demonstrate that the composition of GM-CSF and apoptosis-inducing proteins in the VV genome is very efficient tool for specific killing of cancer cells and for activation of antitumor immunity.
Collapse
Affiliation(s)
- Olga Koval
- Department of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Galina Kochneva
- Department of Viral Hepatitis, State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Russia
| | - Anastasiya Tkachenko
- Department of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
| | - Olga Troitskaya
- Department of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Galina Sivolobova
- Department of Viral Hepatitis, State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Russia
| | - Antonina Grazhdantseva
- Department of Viral Hepatitis, State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Russia
| | - Anna Nushtaeva
- Department of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
| | - Elena Kuligina
- Department of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
| | - Vladimir Richter
- Department of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, SB RAS, Novosibirsk, Russia
| |
Collapse
|
16
|
Vuong CN, Chou WK, Kuttappan VA, Hargis BM, Bielke LR, Berghman LR. A Fast and Inexpensive Protocol for Empirical Verification of Neutralizing Epitopes in Microbial Toxins and Enzymes. Front Vet Sci 2017; 4:91. [PMID: 28660200 PMCID: PMC5468450 DOI: 10.3389/fvets.2017.00091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/30/2017] [Indexed: 12/31/2022] Open
Abstract
In vivo targeting of peptides to antigen-presenting cells by use of agonistic anti-CD40 monoclonal antibodies has been used successfully as an immune response enhancing strategy. When tested in chickens, the antibody-guided platform was capable of inducing specific IgG production within 1 week postimmunization. However, use of this method beyond its initial conception as a vaccine delivery tool has not been fully exploited. In this study, Clostridium perfringens alpha-toxin was used as a model microbial toxin for epitope mapping by using the antibody-guided immunization method to generate a panel of antibodies against specific, regions of the toxin in an attempt to identify crucial determinants on the toxin which, once bound, would hinder downstream toxicity. Alpha-toxin, which possesses both hemolytic and phospholipase C (PLC) enzymatic activities, has long been known to be one of the key destructive etiological agents of necrotic enteritis disease in poultry. Previous attempts to identify crucial antigenic determinants on the toxin mediating its enzymatic activities have been performed using expensive and labor-intensive site-directed mutagenesis techniques. To create a panel of antibodies, 23 short candidate alpha-toxin peptide regions were selected in silico using B-cell epitope prediction algorithms in the public domain and were custom synthesized to load onto the antibody-guided complex for immunization in birds for antisera production. Peptide-specific antibody responses were generated against all candidate neutralizing epitopes and used for in vitro toxin neutralization tests. Antisera against all 23 peptides were able to neutralize the toxin’s hemolytic activity, with neutralization titers ranging from 80 to 320, but none were effective in blocking PLC. The novel approach of antibody-guided immunization introduces a new, inexpensive method for polyclonal IgG production and de facto identification of neutralizing epitopes in microbial toxins and enzymes within 2 weeks from in silico analysis of a putative target sequence.
Collapse
Affiliation(s)
- Christine N Vuong
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Wen-Ko Chou
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
| | - Vivek A Kuttappan
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Billy M Hargis
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Lisa R Bielke
- Department of Animal Science, The Ohio State University, Columbus, OH, United States
| | - Luc R Berghman
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States.,Department of Poultry Science, Texas A&M University, College Station, TX, United States
| |
Collapse
|
17
|
Kilinc MO, Ehrig K, Pessian M, Minev BR, Szalay AA. Colonization of xenograft tumors by oncolytic vaccinia virus (VACV) results in enhanced tumor killing due to the involvement of myeloid cells. J Transl Med 2016; 14:340. [PMID: 27993141 PMCID: PMC5168712 DOI: 10.1186/s12967-016-1096-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/24/2016] [Indexed: 12/25/2022] Open
Abstract
Background The mechanisms by which vaccinia virus (VACV) interacts with the innate immune components are complex and involve different mechanisms. iNOS-mediated NO production by myeloid cells is one of the central antiviral mechanisms and this study aims to investigate specifically whether iNOS-mediated NO production by myeloid cells, is involved in tumor eradication following the virus treatment. Methods Human colon adenocarcinoma (HCT-116) xenograft tumors were infected by VACV. Infiltration of iNOS+ myeloid cell population into the tumor, and virus titer was monitored following the treatment. Single-cell suspensions were stained for qualitative and quantitative flow analysis. The effect of different myeloid cell subsets on tumor growth and colonization were investigated by depletion studies. Finally, in vitro culture experiments were carried out to study NO production and tumor cell killing. Student’s t test was used for comparison between groups in all of the experiments. Results Infection of human colon adenocarcinoma (HCT-116) xenograft tumors by VACV has led to recruitment of many CD11b+ ly6G+ myeloid-derived suppressor cells (MDSCs), with enhanced iNOS expression in the tumors, and to an increased intratumoral virus titer between days 7 and 10 post-VACV therapy. In parallel, both single and multiple rounds of iNOS-producing cell depletions caused very rapid tumor growth within the same period after virus injection, indicating that VACV-induced iNOS+ MDSCs could be an important antitumor effector component. A continuous blockade of iNOS by its specific inhibitor, L-NIL, showed similar tumor growth enhancement 7–10 days post-infection. Finally, spleen-derived iNOS+ MDSCs isolated from virus-injected tumor bearing mice produced higher amounts of NO and effectively killed HCT-116 cells in in vitro transwell experiments. Conclusions We initially hypothesized that NO could be one of the factors that limits active spreading of the virus in the cancerous tissue. In contrast to our initial hypothesis, we observed that PMN-MDSCs were the main producer of NO through iNOS and NO provided a beneficial antitumor effect, The results strongly support an important novel role for VACV infection in the tumor microenvironment. VACV convert tumor-promoting MDSCs into tumor-killing cells by inducing higher NO production.
Collapse
Affiliation(s)
- Mehmet Okyay Kilinc
- Department of Biochemistry, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany.,San Diego Science Center, Genelux Corporation, San Diego, CA, USA
| | - Klaas Ehrig
- Department of Biochemistry, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Maysam Pessian
- San Diego Science Center, Genelux Corporation, San Diego, CA, USA
| | - Boris R Minev
- San Diego Science Center, Genelux Corporation, San Diego, CA, USA.,Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA, USA
| | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany. .,San Diego Science Center, Genelux Corporation, San Diego, CA, USA. .,Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA, USA.
| |
Collapse
|
18
|
Choi AH, O'Leary MP, Fong Y, Chen NG. From Benchtop to Bedside: A Review of Oncolytic Virotherapy. Biomedicines 2016; 4:biomedicines4030018. [PMID: 28536385 PMCID: PMC5344257 DOI: 10.3390/biomedicines4030018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) demonstrate the ability to replicate selectively in cancer cells, resulting in antitumor effects by a variety of mechanisms, including direct cell lysis and indirect cell death through immune-mediate host responses. Although the mechanisms of action of OVs are still not fully understood, major advances have been made in our understanding of how OVs function and interact with the host immune system, resulting in the recent FDA approval of the first OV for cancer therapy in the USA. This review provides an overview of the history of OVs, their selectivity for cancer cells, and their multifaceted mechanism of antitumor action, as well as strategies employed to augment selectivity and efficacy of OVs. OVs in combination with standard cancer therapies are also discussed, as well as a review of ongoing human clinical trials.
Collapse
Affiliation(s)
- Audrey H Choi
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Michael P O'Leary
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Nanhai G Chen
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
19
|
Hirvinen M, Capasso C, Guse K, Garofalo M, Vitale A, Ahonen M, Kuryk L, Vähä-Koskela M, Hemminki A, Fortino V, Greco D, Cerullo V. Expression of DAI by an oncolytic vaccinia virus boosts the immunogenicity of the virus and enhances antitumor immunity. MOLECULAR THERAPY-ONCOLYTICS 2016; 3:16002. [PMID: 27626058 PMCID: PMC5008257 DOI: 10.1038/mto.2016.2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 12/18/2015] [Indexed: 12/17/2022]
Abstract
In oncolytic virotherapy, the ability of the virus to activate the immune system is a key attribute with regard to long-term antitumor effects. Vaccinia viruses bear one of the strongest oncolytic activities among all oncolytic viruses. However, its capacity for stimulation of antitumor immunity is not optimal, mainly due to its immunosuppressive nature. To overcome this problem, we developed an oncolytic VV that expresses intracellular pattern recognition receptor DNA-dependent activator of IFN-regulatory factors (DAI) to boost the innate immune system and to activate adaptive immune cells in the tumor. We showed that infection with DAI-expressing VV increases expression of several genes related to important immunological pathways. Treatment with DAI-armed VV resulted in significant reduction in the size of syngeneic melanoma tumors in mice. When the mice were rechallenged with the same tumor, DAI-VV-treated mice completely rejected growth of the new tumor, which indicates immunity established against the tumor. We also showed enhanced control of growth of human melanoma tumors and elevated levels of human T-cells in DAI-VV-treated mice humanized with human peripheral blood mononuclear cells. We conclude that expression of DAI by an oncolytic VV is a promising way to amplify the vaccine potency of an oncolytic vaccinia virus to trigger the innate-and eventually the long-lasting adaptive immunity against cancer.
Collapse
Affiliation(s)
- Mari Hirvinen
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| | - Cristian Capasso
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| | - Kilian Guse
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki , Helsinki, Finland
| | - Mariangela Garofalo
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| | - Andrea Vitale
- Department of Movement Sciences and Wellness (DiSMEB), University of Naples Parthenope and CEINGE-Biotecnologie Avanzate , Naples, Italy
| | - Marko Ahonen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki , Helsinki, Finland
| | - Lukasz Kuryk
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Oncos Therapeutics Ltd., Helsinki, Finland
| | | | - Akseli Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics, Ltd., Helsinki, Finland; Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Dario Greco
- Finnish Institute of Occupational Health , Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| |
Collapse
|
20
|
Pol J, Buqué A, Aranda F, Bloy N, Cremer I, Eggermont A, Erbs P, Fucikova J, Galon J, Limacher JM, Preville X, Sautès-Fridman C, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch-Oncolytic viruses and cancer therapy. Oncoimmunology 2016; 5:e1117740. [PMID: 27057469 PMCID: PMC4801444 DOI: 10.1080/2162402x.2015.1117740] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 02/06/2023] Open
Abstract
Oncolytic virotherapy relies on the administration of non-pathogenic viral strains that selectively infect and kill malignant cells while favoring the elicitation of a therapeutically relevant tumor-targeting immune response. During the past few years, great efforts have been dedicated to the development of oncolytic viruses with improved specificity and potency. Such an intense wave of investigation has culminated this year in the regulatory approval by the US Food and Drug Administration (FDA) of a genetically engineered oncolytic viral strain for use in melanoma patients. Here, we summarize recent preclinical and clinical advances in oncolytic virotherapy.
Collapse
Affiliation(s)
- Jonathan Pol
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Aitziber Buqué
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Norma Bloy
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Isabelle Cremer
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | | | | | - Jitka Fucikova
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers, Paris, France
| | | | | | - Catherine Sautès-Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | - Radek Spisek
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, U1015, CICBT507, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
21
|
Tsun A, Miao XN, Wang CM, Yu DC. Oncolytic Immunotherapy for Treatment of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:241-83. [PMID: 27240460 DOI: 10.1007/978-94-017-7555-7_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Immunotherapy entails the treatment of disease by modulation of the immune system. As detailed in the previous chapters, the different modes of achieving immune modulation are many, including the use of small/large molecules, cellular therapy, and radiation. Oncolytic viruses that can specifically attack, replicate within, and destroy tumors represent one of the most promising classes of agents for cancer immunotherapy (recently termed as oncolytic immunotherapy). The notion of oncolytic immunotherapy is considered as the way in which virus-induced tumor cell death (known as immunogenic cancer cell death (ICD)) allows the immune system to recognize tumor cells and provide long-lasting antitumor immunity. Both immune responses toward the virus and ICD together contribute toward successful antitumor efficacy. What is now becoming increasingly clear is that monotherapies, through any of the modalities detailed in this book, are neither sufficient in eradicating tumors nor in providing long-lasting antitumor immune responses and that combination therapies may deliver enhanced efficacy. After the rise of the genetic engineering era, it has been possible to engineer viruses to harbor combination-like characteristics to enhance their potency in cancer immunotherapy. This chapter provides a historical background on oncolytic virotherapy and its future application in cancer immunotherapy, especially as a combination therapy with other treatment modalities.
Collapse
Affiliation(s)
- A Tsun
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - X N Miao
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - C M Wang
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - D C Yu
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China.
| |
Collapse
|
22
|
|
23
|
García-Arriaza J, Esteban M. Enhancing poxvirus vectors vaccine immunogenicity. Hum Vaccin Immunother 2015; 10:2235-44. [PMID: 25424927 DOI: 10.4161/hv.28974] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Attenuated recombinant poxvirus vectors expressing heterologous antigens from pathogens are currently at various stages in clinical trials with the aim to establish their efficacy. This is because these vectors have shown excellent safety profiles, significant immunogenicity against foreign expressed antigens and are able to induce protective immune responses. In view of the limited efficacy triggered by some poxvirus strains used in clinical trials (i.e, ALVAC in the RV144 phase III clinical trial for HIV), and of the restrictive replication capacity of the highly attenuated vectors like MVA and NYVAC, there is a consensus that further improvements of these vectors should be pursuit. In this review we considered several strategies that are currently being implemented, as well as new approaches, to improve the immunogenicity of the poxvirus vectors. This includes heterologous prime/boost protocols, use of co-stimulatory molecules, deletion of viral immunomodulatory genes still present in the poxvirus genome, enhancing virus promoter strength, enhancing vector replication capacity, optimizing expression of foreign heterologous sequences, and the combined use of adjuvants. An optimized poxvirus vector triggering long-lasting immunity with a high protective efficacy against a selective disease should be sought.
Collapse
Affiliation(s)
- Juan García-Arriaza
- a Department of Molecular and Cellular Biology; Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| | | |
Collapse
|
24
|
Sánchez-Sampedro L, Perdiguero B, Mejías-Pérez E, García-Arriaza J, Di Pilato M, Esteban M. The evolution of poxvirus vaccines. Viruses 2015; 7:1726-803. [PMID: 25853483 PMCID: PMC4411676 DOI: 10.3390/v7041726] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
After Edward Jenner established human vaccination over 200 years ago, attenuated poxviruses became key players to contain the deadliest virus of its own family: Variola virus (VARV), the causative agent of smallpox. Cowpox virus (CPXV) and horsepox virus (HSPV) were extensively used to this end, passaged in cattle and humans until the appearance of vaccinia virus (VACV), which was used in the final campaigns aimed to eradicate the disease, an endeavor that was accomplished by the World Health Organization (WHO) in 1980. Ever since, naturally evolved strains used for vaccination were introduced into research laboratories where VACV and other poxviruses with improved safety profiles were generated. Recombinant DNA technology along with the DNA genome features of this virus family allowed the generation of vaccines against heterologous diseases, and the specific insertion and deletion of poxvirus genes generated an even broader spectrum of modified viruses with new properties that increase their immunogenicity and safety profile as vaccine vectors. In this review, we highlight the evolution of poxvirus vaccines, from first generation to the current status, pointing out how different vaccines have emerged and approaches that are being followed up in the development of more rational vaccines against a wide range of diseases.
Collapse
MESH Headings
- Animals
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Poxviridae/immunology
- Poxviridae/isolation & purification
- Smallpox/prevention & control
- Smallpox Vaccine/history
- Smallpox Vaccine/immunology
- Smallpox Vaccine/isolation & purification
- Vaccines, Attenuated/history
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/history
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
Collapse
Affiliation(s)
- Lucas Sánchez-Sampedro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Mauro Di Pilato
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| |
Collapse
|
25
|
Parviainen S, Autio K, Vähä-Koskela M, Guse K, Pesonen S, Rosol TJ, Zhao F, Hemminki A. Incomplete but infectious vaccinia virions are produced in the absence of oncolysis in feline SCCF1 cells. PLoS One 2015; 10:e0120496. [PMID: 25799430 PMCID: PMC4370597 DOI: 10.1371/journal.pone.0120496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/23/2015] [Indexed: 12/03/2022] Open
Abstract
Vaccinia virus is a large, enveloped virus of the poxvirus family. It has broad tropism and typically virus replication culminates in accumulation and lytic release of intracellular mature virus (IMV), the most abundant form of infectious virus, as well as release by budding of extracellular enveloped virus (EEV). Vaccinia viruses have been modified to replicate selectively in cancer cells and clinically tested as oncolytic agents. During preclinical screening of relevant cancer targets for a recombinant Western Reserve strain deleted for both copies of the thymidine kinase and vaccinia growth factor genes, we noticed that confluent monolayers of SCCF1 cat squamous carcinoma cells were not destroyed even after prolonged infection. Interestingly, although SCCF1 cells were not killed, they continuously secreted virus into the cell culture supernatant. To investigate this finding further, we performed detailed studies by electron microscopy. Both intracellular and secreted virions showed morphological abnormalities on ultrastructural inspection, suggesting compromised maturation and morphogenesis of vaccinia virus in SCCF1 cells. Our data suggest that SCCF1 cells produce a morphologically abnormal virus which is nevertheless infective, providing new information on the virus-host cell interactions and intracellular biology of vaccinia virus.
Collapse
Affiliation(s)
- Suvi Parviainen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Karoliina Autio
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Kilian Guse
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Sari Pesonen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Thomas J. Rosol
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, The United States of America
| | - Fang Zhao
- Advanced Microscopy Unit, Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
26
|
Hirvinen M, Rajecki M, Kapanen M, Parviainen S, Rouvinen-Lagerström N, Diaconu I, Nokisalmi P, Tenhunen M, Hemminki A, Cerullo V. Immunological effects of a tumor necrosis factor alpha-armed oncolytic adenovirus. Hum Gene Ther 2015; 26:134-44. [PMID: 25557131 DOI: 10.1089/hum.2014.069] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
For long it has been recognized that tumor necrosis factor alpha (TNFa) has anticancer characteristics, and its use as a cancer therapeutic was proposed already in the 1980s. However, its systemic toxicity has limited its usability. Oncolytic viruses, selectively cancer-killing viruses, have shown great potency, and one of their most useful aspects is their ability to produce high amounts of transgene products locally, resulting in high local versus systemic concentrations. Therefore, the overall magnitude of tumor cell killing results from the combination of oncolysis, transgene-mediated direct effect such as TNFa-mediated apoptosis, and, perhaps most significantly, from activation of the host immune system against the tumor. We generated a novel chimeric oncolytic adenovirus expressing human TNFa, Ad5/3-D24-hTNFa, whose efficacy and immunogenicity were tested in vitro and in vivo. The hTNFa-expressing adenovirus showed increased cancer-eradicating potency, which was shown to be because of elevated apoptosis and necrosis rates and induction of various immune responses. Interestingly, we saw increase in immunogenic cell death markers in Ad5/3-d24-hTNFa-treated cells. Moreover, tumors treated with Ad5/3-D24-hTNFa displayed enhanced presence of OVA-specific cytotoxic T cells. We thus can conclude that tumor eradication and antitumor immune responses mediated by Ad5/3-d24-hTNFa offer a new potential drug candidate for cancer therapy.
Collapse
Affiliation(s)
- Mari Hirvinen
- 1 Laboratory of Immunovirotherapy, Division of Pharmaceutical Biosciences and Centre for Drug Research, Faculty of Pharmacy, University of Helsinki , 00790 Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Overcoming tumor resistance by heterologous adeno-poxvirus combination therapy. MOLECULAR THERAPY-ONCOLYTICS 2015; 1:14006. [PMID: 27119097 PMCID: PMC4782942 DOI: 10.1038/mto.2014.6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 09/02/2014] [Indexed: 12/17/2022]
Abstract
Successful cancer control relies on overcoming resistance to cell death and on activation of host antitumor immunity. Oncolytic viruses are particularly attractive in this regard, as they lyse infected tumor cells and trigger robust immune responses during the infection. However, repeated injections of the same virus promote antiviral rather than antitumor immunity and tumors may mount innate antiviral defenses to restrict oncolytic virus replication. In this article, we have explored if alternating the therapy virus could circumvent these problems. We demonstrate in two virus-resistant animal models a substantial delay in antiviral immune- and innate cellular response induction by alternating injections of two immunologically distinct oncolytic viruses, adenovirus, and vaccinia virus. Our results are in support of clinical development of heterologous adeno-/vaccinia virus therapy of cancer.
Collapse
|
28
|
Zamarin D, Wolchok JD. Potentiation of immunomodulatory antibody therapy with oncolytic viruses for treatment of cancer. MOLECULAR THERAPY-ONCOLYTICS 2014; 1:14004. [PMID: 27119094 PMCID: PMC4782939 DOI: 10.1038/mto.2014.4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 02/07/2023]
Abstract
Identification of the immune suppressive mechanisms active within the tumor microenvironment led to development of immunotherapeutic strategies aiming to reverse the immunosuppression and to enhance the function of tumor-infiltrating lymphocytes. Of those, cancer therapy with antibodies targeting the immune costimulatory and coinhibitory receptors has demonstrated significant promise in the recent years, with multiple antibodies entering clinical testing. The responses to these agents, however, have not been universal and have not been observed in all cancer types, calling for identification of appropriate predictive biomarkers and development of combinatorial strategies. Pre-existing immune infiltration in tumors has been demonstrated to have a strong association with response to immunotherapies, with the type I interferon (IFN) pathway emerging as a key player in tumor innate immune recognition and activation of adaptive immunity. These findings provide a rationale for evaluation of strategies targeting the type I IFN pathway as a means to enhance tumor immune recognition and infiltration, which could potentially make them susceptible to therapeutics targeting the cosignaling receptors. To this end in particular, oncolytic viruses (OVs) have been demonstrated to enhance tumor recognition by the immune system through multiple mechanisms, which include upregulation of major histocompatibility complex and costimulatory molecules on cancer cells, immunogenic cell death and antigen release, and activation of the type I IFN pathway. Evidence is now emerging that combination therapies using OVs and agents targeting immune cosignaling receptors such as 4-1BB, PD-1, and CTLA-4 may work in concert to enhance antitumor immunity and therapeutic efficacy. Our evolving understanding of the interplay between OVs and the immune system demonstrates that the virus-induced antitumor immune responses can be harnessed to drive the efficacy of the agents targeting cosignaling receptors and provides a strong rationale for integration of such therapies in clinic.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA; Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University, New York, New York, USA
| | - Jedd D Wolchok
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, USA; Department of Medicine, Melanoma and Immunotherapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, New York, USA; Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University, New York, New York, USA
| |
Collapse
|
29
|
Autio K, Knuuttila A, Kipar A, Pesonen S, Guse K, Parviainen S, Rajamäki M, Laitinen-Vapaavuori O, Vähä-Koskela M, Kanerva A, Hemminki A. Safety and biodistribution of a double-deleted oncolytic vaccinia virus encoding CD40 ligand in laboratory Beagles. Mol Ther Oncolytics 2014; 1:14002. [PMID: 27119092 PMCID: PMC4782937 DOI: 10.1038/mto.2014.2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/26/2014] [Indexed: 12/13/2022] Open
Abstract
We evaluated adverse events, biodistribution and shedding of oncolytic vaccinia virus encoding CD40 ligand in two Beagles, in preparation for a phase 1 trial in canine cancer patients. Dog 1 received one dose of vaccinia virus and was euthanized 24 hours afterwards, while dog 2 received virus four times once weekly and was euthanized 7 days after that. Dogs were monitored for adverse events and underwent a detailed postmortem examination. Blood, saliva, urine, feces, and organs were collected for virus detection. Dog 1 had mild fever and lethargy while dog 2 experienced a possible seizure 5.5 hours after first virus administration. Viral DNA declined quickly in the blood after virus administration in both dogs but was still detectable 1 week later by quantitative polymerase chain reaction. Only samples taken directly after virus infusion contained infectious virus. Small amounts of viral DNA, but no infectious virus, were detected in a few saliva and urine samples. Necropsies did not reveal any relevant pathological changes and virus DNA was detected mainly in the spleen. The dogs in the study did not have cancer, and thus adverse events could be more common and viral load higher in dogs with tumors which allow viral amplification.
Collapse
Affiliation(s)
- Karoliina Autio
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Anna Knuuttila
- Finnish Centre for Laboratory Animal Pathology and Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Anja Kipar
- Finnish Centre for Laboratory Animal Pathology and Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sari Pesonen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Kilian Guse
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Suvi Parviainen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Minna Rajamäki
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Outi Laitinen-Vapaavuori
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| |
Collapse
|
30
|
Autio K, Knuuttila A, Kipar A, Ahonen M, Parviainen S, Diaconu I, Kanerva A, Hakonen T, Vähä-Koskela M, Hemminki A. Anti-tumour activity of oncolytic Western Reserve vaccinia viruses in canine tumour cell lines, xenografts, and fresh tumour biopsies. Vet Comp Oncol 2014; 14:395-408. [PMID: 25302859 DOI: 10.1111/vco.12119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 08/29/2014] [Accepted: 09/09/2014] [Indexed: 12/13/2022]
Abstract
Cancer is one of the most common reasons for death in dogs. One promising approach is oncolytic virotherapy. We assessed the oncolytic effect of genetically modified vaccinia viruses in canine cancer cells, in freshly excised tumour biopsies, and in mice harbouring canine tumour xenografts. Tumour transduction efficacy was assessed using virus expressing luciferase or fluorescent marker genes and oncolysis was quantified by a colorimetric cell viability assay. Oncolytic efficacy in vivo was evaluated in a nude mouse xenograft model. Vaccinia virus was shown to infect most tested canine cancer cell lines and primary surgical tumour tissues. Virus infection significantly reduced tumour growth in the xenograft model. Oncolytic vaccinia virus has antitumour effects against canine cancer cells and experimental tumours and is able to replicate in freshly excised patient tumour tissue. Our results suggest that oncolytic vaccinia virus may offer an effective treatment option for otherwise incurable canine tumours.
Collapse
Affiliation(s)
- K Autio
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.,Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - A Knuuttila
- Finnish Centre for Laboratory Animal Pathology and Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - A Kipar
- Finnish Centre for Laboratory Animal Pathology and Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - M Ahonen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - S Parviainen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - I Diaconu
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - A Kanerva
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland.,Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - T Hakonen
- Oncos Therapeutics Ltd, Helsinki, Finland
| | - M Vähä-Koskela
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - A Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland
| |
Collapse
|
31
|
Parviainen S, Ahonen M, Diaconu I, Kipar A, Siurala M, Vähä-Koskela M, Kanerva A, Cerullo V, Hemminki A. GMCSF-armed vaccinia virus induces an antitumor immune response. Int J Cancer 2014; 136:1065-72. [PMID: 25042001 DOI: 10.1002/ijc.29068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/23/2014] [Indexed: 12/14/2022]
Abstract
Oncolytic Western Reserve strain vaccinia virus selective for epidermal growth factor receptor pathway mutations and tumor-associated hypermetabolism was armed with human granulocyte-macrophage colony-stimulating factor (GMCSF) and a tdTomato fluorophore. As the assessment of immunological responses to human transgenes is challenging in the most commonly used animal models, we used immunocompetent Syrian golden hamsters, known to be sensitive to human GMCSF and semipermissive to vaccinia virus. Efficacy was initially tested in vitro on various human and hamster cell lines and oncolytic potency of transgene-carrying viruses was similar to unarmed virus. The hGMCSF-encoding virus was able to completely eradicate subcutaneous pancreatic tumors in hamsters, and to fully protect the animals from subsequent rechallenge with the same tumor. Induction of specific antitumor immunity was also shown by ex vivo co-culture experiments with hamster splenocytes. In addition, histological examination revealed increased infiltration of neutrophils and macrophages in GMCSF-virus-treated tumors. These findings help clarify the mechanism of action of GMCSF-armed vaccinia viruses undergoing clinical trials.
Collapse
Affiliation(s)
- Suvi Parviainen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Cremer I, Erbs P, Limacher JM, Preville X, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch:: Oncolytic viruses for cancer therapy. Oncoimmunology 2014; 3:e28694. [PMID: 25097804 PMCID: PMC4091053 DOI: 10.4161/onci.28694] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022] Open
Abstract
Oncolytic viruses are natural or genetically modified viral species that selectively infect and kill neoplastic cells. Such an innate or exogenously conferred specificity has generated considerable interest around the possibility to employ oncolytic viruses as highly targeted agents that would mediate cancer cell-autonomous anticancer effects. Accumulating evidence, however, suggests that the therapeutic potential of oncolytic virotherapy is not a simple consequence of the cytopathic effect, but strongly relies on the induction of an endogenous immune response against transformed cells. In line with this notion, superior anticancer effects are being observed when oncolytic viruses are engineered to express (or co-administered with) immunostimulatory molecules. Although multiple studies have shown that oncolytic viruses are well tolerated by cancer patients, the full-blown therapeutic potential of oncolytic virotherapy, especially when implemented in the absence of immunostimulatory interventions, remains unclear. Here, we cover the latest advances in this active area of translational investigation, summarizing high-impact studies that have been published during the last 12 months and discussing clinical trials that have been initiated in the same period to assess the therapeutic potential of oncolytic virotherapy in oncological indications.
Collapse
Affiliation(s)
- Jonathan Pol
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Norma Bloy
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | | | | | | | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
| |
Collapse
|