851
|
Lanoie D, Lemay G. Multiple proteins differing between laboratory stocks of mammalian orthoreoviruses affect both virus sensitivity to interferon and induction of interferon production during infection. Virus Res 2018; 247:40-46. [PMID: 29382551 DOI: 10.1016/j.virusres.2018.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/20/2018] [Accepted: 01/21/2018] [Indexed: 12/19/2022]
Abstract
In the course of previous works, it was observed that the virus laboratory stock (T3DS) differs in sequence from the virus encoded by the ten plasmids currently in use in many laboratories (T3DK), and derived from a different original virus stock. Seven proteins are affected by these sequence differences. In the present study, replication of T3DK was shown to be more sensitive to the antiviral effect of interferon. Infection by the T3DK virus was also shown to induce the production of higher amount of β and α-interferons compared to T3DS. Two proteins, the μ2 and λ2 proteins, were found to be responsible for increased sensitivity to interferon while both μ2 and λ1 are responsible for increased interferon secretion. Altogether this supports the idea that multiple reovirus proteins are involved in the control of induction of interferon and virus sensitivity to the interferon-induced response. While interrelated, interferon induction and sensitivity can be separated by defined gene combinations. While both μ2 and λ2 were previously suspected of a role in the control of the interferon response, other proteins are also likely involved, as first shown here for λ1. This also further stresses that due caution should be exerted when comparing different virus isolates with different genetic background.
Collapse
Affiliation(s)
- Delphine Lanoie
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, H3C 3J7, Canada
| | - Guy Lemay
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, H3C 3J7, Canada.
| |
Collapse
|
852
|
Xu Z, Li P, Fan L, Wu M. The Potential Role of circRNA in Tumor Immunity Regulation and Immunotherapy. Front Immunol 2018; 9:9. [PMID: 29403493 PMCID: PMC5786515 DOI: 10.3389/fimmu.2018.00009] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022] Open
Abstract
Non-coding RNAs (ncRNAs) can be divided into circular non-coding RNAs (circRNAs) and linear ncRNAs. ncRNAs exist in different cell types, including normal cells, tumor cells and immunocytes. Linear ncRNAs, such as long ncRNAs and microRNAs, have been found to play important roles in the regulation of tumor immunity and immunotherapy; however, the functions of circRNAs in tumor immunity and immunotherapy are less known. Here, we review the current status of ncRNAs in the regulation of tumor immunity and immunotherapy and emphatically discuss the potential roles of circRNAs as tumor antigens in the regulation of tumor immunity and immunotherapy.
Collapse
Affiliation(s)
- Zihao Xu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Cancer Research Institute, Central South University, Changsha, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Cancer Research Institute, Central South University, Changsha, China
| | - Li Fan
- Department of Biochemistry, University of California Riverside, Riverside, CA, United States
| | - Minghua Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Cancer Research Institute, Central South University, Changsha, China.,Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| |
Collapse
|
853
|
Xie YQ, Wei L, Tang L. Immunoengineering with biomaterials for enhanced cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1506. [PMID: 29333729 DOI: 10.1002/wnan.1506] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/07/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has recently shown dramatic clinical success inducing durable response in patients of a wide variety of malignancies. Further improvement of the clinical outcome with immune related cancer treatment requests more exquisite manipulation of a patient's immune system with increased immunity against diseases while mitigating the toxicities. To meet this challenge, biomaterials applied to immunoengineering are being developed to achieve tissue- and/or cell-specific immunomodulation and thus could potentially enhance both the efficacy and safety of current cancer immunotherapies. Here, we review the recent advancement in the field of immunoengineering using biomaterials and their applications in promoting different modalities of cancer immunotherapies, with focus on cell-, antibody-, immunomodulator-, and gene-based immune related treatments and their combinations with conventional therapies. Challenges and opportunities are discussed in applying biomaterials engineering strategies in the development of future cancer immunotherapies. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- Yu-Qing Xie
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lixia Wei
- Institute of Materials Science & Engineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Institute of Materials Science & Engineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
854
|
Marin-Acevedo JA, Soyano AE, Dholaria B, Knutson KL, Lou Y. Cancer immunotherapy beyond immune checkpoint inhibitors. J Hematol Oncol 2018; 11:8. [PMID: 29329556 PMCID: PMC5767051 DOI: 10.1186/s13045-017-0552-6] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/28/2017] [Indexed: 12/17/2022] Open
Abstract
Malignant cells have the capacity to rapidly grow exponentially and spread in part by suppressing, evading, and exploiting the host immune system. Immunotherapy is a form of oncologic treatment directed towards enhancing the host immune system against cancer. In recent years, manipulation of immune checkpoints or pathways has emerged as an important and effective form of immunotherapy. Agents that target cytotoxic T lymphocyte-associated molecule-4 (CTLA-4), programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1 (PD-L1) are the most widely studied and recognized. Immunotherapy, however, extends beyond immune checkpoint therapy by using new molecules such as chimeric monoclonal antibodies and antibody drug conjugates that target malignant cells and promote their destruction. Genetically modified T cells expressing chimeric antigen receptors are able to recognize specific antigens on cancer cells and subsequently activate the immune system. Native or genetically modified viruses with oncolytic activity are of great interest as, besides destroying malignant cells, they can increase anti-tumor activity in response to the release of new antigens and danger signals as a result of infection and tumor cell lysis. Vaccines are also being explored, either in the form of autologous or allogenic tumor peptide antigens, genetically modified dendritic cells that express tumor peptides, or even in the use of RNA, DNA, bacteria, or virus as vectors of specific tumor markers. Most of these agents are yet under development, but they promise to be important options to boost the host immune system to control and eliminate malignancy. In this review, we have provided detailed discussion of different forms of immunotherapy agents other than checkpoint-modifying drugs. The specific focus of this manuscript is to include first-in-human phase I and phase I/II clinical trials intended to allow the identification of those drugs that most likely will continue to develop and possibly join the immunotherapeutic arsenal in a near future.
Collapse
Affiliation(s)
| | - Aixa E Soyano
- Department of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Bhagirathbhai Dholaria
- Department of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Current address: Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
| | - Yanyan Lou
- Department of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
855
|
Viruses, bacteria, and parasites - oh my! a resurgence of interest in microbial-based therapy for cancer. J Immunother Cancer 2018; 6:3. [PMID: 29307306 PMCID: PMC5757293 DOI: 10.1186/s40425-017-0312-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/18/2017] [Indexed: 11/22/2022] Open
Abstract
As infections and cancer are two of the most common maladies affecting human beings, a concerted effort is needed to better understand their potential interactions and to further explore their use in microbial-based cancer treatments. Studies focusing on the interaction between pathogens and cancer began over 4000 years ago, but therapeutic application of pathogens has often been bypassed as other cancer therapies have gained wider interest. To many, the field of microbial-based cancer treatment may feel antiquated and already sufficiently explored. However, closer examination reveals that our current knowledge is but a series of dim reflections amongst many yet-unexplored shadows. Particularly, with our increased understanding of pathogen entry, replication, and senescence, coupled with our quickly increasing knowledge regarding cancer initiation, growth, and metastasis, and capped by our realization of the complexity and plasticity of the immune response, we are just now beginning to realize the vastness of the undiscovered area encompassing this field. At the same time, we are now uniquely poised with gained knowledge and discovered tools to join together across disciplines, uncover new positive and negative interactions between pathogens and cancer, and make important progress toward saving cancer patient lives.
Collapse
|
856
|
Taipale K, Tähtinen S, Havunen R, Koski A, Liikanen I, Pakarinen P, Koivisto-Korander R, Kankainen M, Joensuu T, Kanerva A, Hemminki A. Interleukin 8 activity influences the efficacy of adenoviral oncolytic immunotherapy in cancer patients. Oncotarget 2018; 9:6320-6335. [PMID: 29464075 PMCID: PMC5814215 DOI: 10.18632/oncotarget.23967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 12/12/2017] [Indexed: 12/20/2022] Open
Abstract
After the landmark approval of T-VEC, oncolytic viruses are finding their way to the clinics. However, response rates have still room for improvement, and unfortunately there are currently no available markers to predict responses for oncolytic immunotherapy. Interleukin 8 (IL-8) production is upregulated in many cancers and it also connects to several pathways that have been shown to impair the efficacy of adenoviral immunotherapy. We studied the role of IL-8 in 103 cancer patients treated with oncolytic adenoviruses. We found high baseline serum IL-8 concentration to be independently associated with poor prognosis (p<0.001). Further, normal baseline IL-8 was associated with improved prognostic potential of calculation of the neutrophil-to-lymphocyte ratio (p<0.001). Interestingly, a decrease in IL-8 concentration after treatment with oncolytic adenovirus predicted better overall survival (p<0.001) and higher response rate, although this difference was not significant (p=0.066). We studied the combination of adenovirus and IL-8 neutralizing antibody ex vivo in single cell suspensions and in co-cultures of tumor-associated CD15+ neutrophils and CD3+ tumor-infiltrating lymphocytes derived from fresh patient tumor samples. These results indicate a role for IL-8 as a biomarker in oncolytic virotherapy, but additionally provide a rationale for targeting IL-8 to improve treatment efficacy. In conclusion, curtailing the activity of IL-8 systemically or locally in the tumor microenvironment could improve anti-tumor immune responses resulting in enhanced efficacy of adenoviral immunotherapy of cancer.
Collapse
Affiliation(s)
- Kristian Taipale
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Siri Tähtinen
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Anniina Koski
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland.,Department of Neurosurgery, HUCH, Helsinki, Finland
| | - Ilkka Liikanen
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Päivi Pakarinen
- Department of Obstetrics and Gynecology, HUCH, Helsinki, Finland
| | | | - Matti Kankainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | | | - Anna Kanerva
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland.,Department of Obstetrics and Gynecology, HUCH, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, University of Helsinki, Faculty of Medicine, Helsinki, Finland.,Docrates Cancer Center, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland.,Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
857
|
Samson A, Scott KJ, Taggart D, West EJ, Wilson E, Nuovo GJ, Thomson S, Corns R, Mathew RK, Fuller MJ, Kottke TJ, Thompson JM, Ilett EJ, Cockle JV, van Hille P, Sivakumar G, Polson ES, Turnbull SJ, Appleton ES, Migneco G, Rose AS, Coffey MC, Beirne DA, Collinson FJ, Ralph C, Alan Anthoney D, Twelves CJ, Furness AJ, Quezada SA, Wurdak H, Errington-Mais F, Pandha H, Harrington KJ, Selby PJ, Vile RG, Griffin SD, Stead LF, Short SC, Melcher AA. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade. Sci Transl Med 2018; 10:eaam7577. [PMID: 29298869 PMCID: PMC6276984 DOI: 10.1126/scitranslmed.aam7577] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 09/08/2017] [Accepted: 11/27/2017] [Indexed: 12/23/2022]
Abstract
Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.
Collapse
Affiliation(s)
- Adel Samson
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK.
| | - Karen J Scott
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - David Taggart
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Emma J West
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Erica Wilson
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Gerard J Nuovo
- Ohio State University, Comprehensive Cancer Centre, Columbus, OH 43210, USA
| | - Simon Thomson
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Robert Corns
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Ryan K Mathew
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Martin J Fuller
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | - Jill M Thompson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Elizabeth J Ilett
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Julia V Cockle
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Philip van Hille
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Gnanamurthy Sivakumar
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Euan S Polson
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Samantha J Turnbull
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Elizabeth S Appleton
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Gemma Migneco
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Ailsa S Rose
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | - Deborah A Beirne
- Leeds Teaching Hospitals National Health Service Trust, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Fiona J Collinson
- Leeds Institute of Clinical Trials Research, Faculty of Medicine and Health, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| | - Christy Ralph
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - D Alan Anthoney
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Christopher J Twelves
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | | | - Heiko Wurdak
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Fiona Errington-Mais
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | | | | | - Peter J Selby
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Richard G Vile
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen D Griffin
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Lucy F Stead
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK
| | - Susan C Short
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK.
| | - Alan A Melcher
- Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK.
| |
Collapse
|
858
|
Sahu SK, Kumar M. Application of Oncolytic Virus as a Therapy of Cancer. Microb Biotechnol 2018. [DOI: 10.1007/978-981-10-7140-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
859
|
Gujar S, Pol JG, Kim Y, Lee PW, Kroemer G. Antitumor Benefits of Antiviral Immunity: An Underappreciated Aspect of Oncolytic Virotherapies. Trends Immunol 2017; 39:209-221. [PMID: 29275092 DOI: 10.1016/j.it.2017.11.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
Oncolytic viruses (OVs) represent a new class of cancer immunotherapeutics. Administration of OVs to cancer-bearing hosts induces two distinct immunities: antiviral and antitumor. While antitumor immunity is beneficial, antiviral immune responses are often considered detrimental for the efficacy of OV-based therapy. The existing dogma postulates that anti-OV immune responses restrict viral replication and spread, and thus reduce direct OV-mediated killing of cancer cells. Accordingly, a myriad of therapeutic strategies aimed at mitigating anti-OV immune responses is presently being tested. Here, we advocate that OV-induced antiviral immune responses hold intrinsic anticancer benefits and are essential for establishing clinically desired antitumor immunity. Thus, to achieve the optimal efficacy of OV-based cancer immunotherapies, strategic management of anti-OV immune responses is of critical importance.
Collapse
Affiliation(s)
- Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Department of Microbiology and Immunology, Dalhousie University, NS, Canada; Department of Biology, Dalhousie University, NS, Canada; Centre for Innovative and Collaborative Health Sciences Research, Quality and System Performance, IWK Health Centre, Halifax, NS, Canada; These authors contributed equally to this work
| | - Jonathan G Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; INSERM, U1138, Paris, 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; Université Pierre et Marie Curie/Paris VI, Paris, France; These authors contributed equally to this work
| | - Youra Kim
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Patrick W Lee
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Department of Microbiology and Immunology, Dalhousie University, NS, Canada; Share senior co-authorship.
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; INSERM, U1138, Paris, 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; Université Pierre et Marie Curie/Paris VI, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden; Share senior co-authorship.
| |
Collapse
|
860
|
Stevens JW, Fletcher C, Downey G, Sutton A. A review of methods for comparing treatments evaluated in studies that form disconnected networks of evidence. Res Synth Methods 2017; 9:148-162. [DOI: 10.1002/jrsm.1278] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 10/04/2017] [Accepted: 10/13/2017] [Indexed: 12/28/2022]
Affiliation(s)
- John W. Stevens
- School of Health and Related Research; University of Sheffield; Regent Court, 30 Regent Street Sheffield UK
| | - Christine Fletcher
- Amgen Ltd, Global Biostatistical Science; 240 Cambridge Science Park, Milton Road Cambridge Cambridgeshire UK
| | - Gerald Downey
- Amgen Ltd, Global Biostatistical Science; 240 Cambridge Science Park, Milton Road Cambridge Cambridgeshire UK
| | - Anthea Sutton
- School of Health and Related Research; University of Sheffield; Regent Court, 30 Regent Street Sheffield UK
| |
Collapse
|
861
|
Angelova AL, Barf M, Geletneky K, Unterberg A, Rommelaere J. Immunotherapeutic Potential of Oncolytic H-1 Parvovirus: Hints of Glioblastoma Microenvironment Conversion towards Immunogenicity. Viruses 2017; 9:v9120382. [PMID: 29244745 PMCID: PMC5744156 DOI: 10.3390/v9120382] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma, one of the most aggressive primary brain tumors, is characterized by highly immunosuppressive microenvironment. This contributes to glioblastoma resistance to standard treatment modalities and allows tumor growth and recurrence. Several immune-targeted approaches have been recently developed and are currently under preclinical and clinical investigation. Oncolytic viruses, including the autonomous protoparvovirus H-1 (H-1PV), show great promise as novel immunotherapeutic tools. In a first phase I/IIa clinical trial (ParvOryx01), H-1PV was safe and well tolerated when locally or systemically administered to recurrent glioblastoma patients. The virus was able to cross the blood-brain (tumor) barrier after intravenous infusion. Importantly, H-1PV treatment of glioblastoma patients was associated with immunogenic changes in the tumor microenvironment. Tumor infiltration with activated cytotoxic T cells, induction of cathepsin B and inducible nitric oxide (NO) synthase (iNOS) expression in tumor-associated microglia/macrophages (TAM), and accumulation of activated TAM in cluster of differentiation (CD) 40 ligand (CD40L)-positive glioblastoma regions was detected. These are the first-in-human observations of H-1PV capacity to switch the immunosuppressed tumor microenvironment towards immunogenicity. Based on this pilot study, we present a tentative model of H-1PV-mediated modulation of glioblastoma microenvironment and propose a combinatorial therapeutic approach taking advantage of H-1PV-induced microglia/macrophage activation for further (pre)clinical testing.
Collapse
Affiliation(s)
- Assia L Angelova
- Department of Tumor Virology (F010), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Milena Barf
- Department of Tumor Virology (F010), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Karsten Geletneky
- Department of Neurosurgery, University Hospital, 69120 Heidelberg, Germany.
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital, 69120 Heidelberg, Germany.
| | - Jean Rommelaere
- Department of Tumor Virology (F010), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
862
|
Potts KG, Irwin CR, Favis NA, Pink DB, Vincent KM, Lewis JD, Moore RB, Hitt MM, Evans DH. Deletion of F4L (ribonucleotide reductase) in vaccinia virus produces a selective oncolytic virus and promotes anti-tumor immunity with superior safety in bladder cancer models. EMBO Mol Med 2017; 9:638-654. [PMID: 28289079 PMCID: PMC5412795 DOI: 10.15252/emmm.201607296] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bladder cancer has a recurrence rate of up to 80% and many patients require multiple treatments that often fail, eventually leading to disease progression. In particular, standard of care for high-grade disease, Bacillus Calmette-Guérin (BCG), fails in 30% of patients. We have generated a novel oncolytic vaccinia virus (VACV) by mutating the F4L gene that encodes the virus homolog of the cell-cycle-regulated small subunit of ribonucleotide reductase (RRM2). The F4L-deleted VACVs are highly attenuated in normal tissues, and since cancer cells commonly express elevated RRM2 levels, have tumor-selective replication and cell killing. These F4L-deleted VACVs replicated selectively in immune-competent rat AY-27 and xenografted human RT112-luc orthotopic bladder cancer models, causing significant tumor regression or complete ablation with no toxicity. It was also observed that rats cured of AY-27 tumors by VACV treatment developed anti-tumor immunity as evidenced by tumor rejection upon challenge and by ex vivo cytotoxic T-lymphocyte assays. Finally, F4L-deleted VACVs replicated in primary human bladder cancer explants. Our findings demonstrate the enhanced safety and selectivity of F4L-deleted VACVs, with application as a promising therapy for patients with BCG-refractory cancers and immune dysregulation.
Collapse
Affiliation(s)
- Kyle G Potts
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Chad R Irwin
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Medical Microbiology & Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nicole A Favis
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Medical Microbiology & Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Desmond B Pink
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Krista M Vincent
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Anatomy & Cell Biology, Faculty of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - John D Lewis
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ronald B Moore
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mary M Hitt
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - David H Evans
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada .,Cancer Research Institute of Northern Alberta (CRINA), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Medical Microbiology & Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
863
|
Oncolytic virotherapy as an immunotherapeutic strategy for multiple myeloma. Blood Cancer J 2017; 7:640. [PMID: 29208938 PMCID: PMC5802552 DOI: 10.1038/s41408-017-0020-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/03/2017] [Accepted: 10/12/2017] [Indexed: 12/22/2022] Open
Abstract
Multiple Myeloma (MM), a clonal malignancy of antibody-producing plasma cells, is the second most common hematologic malignancy and results in significant patient morbidity and mortality. The high degree of immune dysregulation in MM, including T cell imbalances and up-regulation of immunosuppressive checkpoint proteins and myeloid derived suppressor cells, allows this malignancy to escape from host immune control. Despite advances in the therapeutic landscape of MM over the last decade, including the introduction of immunomodulatory drugs, the prognosis for this disease is poor, with less than 50% of patients surviving 5 years. Thus, novel treatment strategies are required. Oncolytic viruses (OV) are a promising new class of therapeutics that rely on tumour specific oncolysis and the generation of a potent adaptive anti-tumour immune response for efficacy. To date, a number of OV have shown efficacy in pre-clinical studies of MM with three reaching early phase clinical trials. OVs represent a rational therapeutic strategy for MM based on (1) their tumour tropism, (2) their ability to potentiate anti-tumour immunity and (3) their ability to be rationally combined with other immunotherapeutic agents to achieve a more robust clinical response.
Collapse
|
864
|
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: 4.6] [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
|
865
|
Choi AH, O'Leary MP, Chaurasiya S, Lu J, Kim SI, Fong Y, Chen NG. Novel chimeric parapoxvirus CF189 as an oncolytic immunotherapy in triple-negative breast cancer. Surgery 2017; 163:336-342. [PMID: 29174433 DOI: 10.1016/j.surg.2017.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/13/2017] [Accepted: 09/16/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Triple-negative breast cancer is an aggressive subtype of breast cancer with high recurrence rate and poor prognosis. Here we describe a novel, genetically engineered parapoxvirus that efficiently kills triple-negative breast cancer. METHODS A novel chimeric parapoxvirus (CF189) was generated via homologous recombination and identified through high-throughput screening. Cytotoxicity was assayed in vitro in 4 triple-negative breast cancer cell lines. Viral replication was examined through standard plaque assay. Orthotopic triple-negative breast cancer xenografts were generated by MDA-MB-468 implantation into the 2nd and 4th mammary fat pads of athymic nude mice and treated with the virus. RESULTS Chimeric parapoxvirus (CF189) demonstrated dose-dependent cytotoxicity at low multiplicity of infection, with > 80% cell death 6 days after treatment. Significant reductions in tumor size were observed 2 weeks after intratumoral injection at doses as low as 103 plaque-forming units (PFU) compared with control (P < 0.01). In addition, abscopal effect (shrinkage of noninjected remote tumors) was clearly demonstrated. CONCLUSION Chimeric parapoxvirus (CF189) demonstrated efficient cytotoxicity in vitro and potent antitumor effect in vivo at doses as low as 103 PFU. These are data encouraging of clinical development for this highly potent agent against triple-negative breast cancer.
Collapse
Affiliation(s)
- Audrey H Choi
- Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Michael P O'Leary
- Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | | | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA; Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA
| | - Nanhai G Chen
- Department of Surgery, City of Hope National Medical Center, Duarte, CA; Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA.
| |
Collapse
|
866
|
Ajina A, Maher J. Prospects for combined use of oncolytic viruses and CAR T-cells. J Immunother Cancer 2017; 5:90. [PMID: 29157300 PMCID: PMC5696728 DOI: 10.1186/s40425-017-0294-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/17/2017] [Indexed: 12/18/2022] Open
Abstract
With the approval of talimogene laherparepvec (T-VEC) for inoperable locally advanced or metastatic malignant melanoma in the USA and Europe, oncolytic virotherapy is now emerging as a viable therapeutic option for cancer patients. In parallel, following the favourable results of several clinical trials, adoptive cell transfer using chimeric antigen receptor (CAR)-redirected T-cells is anticipated to enter routine clinical practice for the management of chemotherapy-refractory B-cell malignancies. However, CAR T-cell therapy for patients with advanced solid tumours has proved far less successful. This Review draws upon recent advances in the design of novel oncolytic viruses and CAR T-cells and provides a comprehensive overview of the synergistic potential of combination oncolytic virotherapy with CAR T-cell adoptive cell transfer for the management of solid tumours, drawing particular attention to the methods by which recombinant oncolytic viruses may augment CAR T-cell trafficking into the tumour microenvironment, mitigate or reverse local immunosuppression and enhance CAR T-cell effector function and persistence.
Collapse
Affiliation(s)
- Adam Ajina
- Department of Oncology, Royal Free London NHS Foundation Trust, London, UK
| | - John Maher
- King’s College London, CAR Mechanics Group, School of Cancer and Pharmaceutical Sciences, Guy’s Hospital Campus, Great Maze Pond, London, SE1 9RT UK
- Department of Clinical Immunology and Allergy, King’s College Hospital NHS Foundation Trust, London, UK
- Department of Immunology, Eastbourne Hospital, East Sussex, UK
| |
Collapse
|
867
|
Margiewicz S, Cordova C, Chi AS, Jain R. State of the Art Treatment and Surveillance Imaging of Glioblastomas. Semin Roentgenol 2017; 53:23-36. [PMID: 29405952 DOI: 10.1053/j.ro.2017.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Christine Cordova
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY
| | - Andrew S Chi
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY
| | - Rajan Jain
- Department of Radiology, NYU School of Medicine, New York, NY; Department of Neurosurgery, NYU School of Medicine, New York, NY.
| |
Collapse
|
868
|
Re-designing Interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent. Nat Commun 2017; 8:1395. [PMID: 29123084 PMCID: PMC5680234 DOI: 10.1038/s41467-017-01385-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022] Open
Abstract
Interleukin-12 (IL-12) has emerged as one of the most potent agents for anti-tumor immunotherapy. However, potentially lethal toxicity associated with systemic administration of IL-12 precludes its clinical application. Here we redesign the molecule in such a way that its anti-tumor efficacy is not compromised, but toxic effects are eliminated. Deletion of the N-terminal signal peptide of IL-12 can effect such a change by preventing IL-12 secretion from cells. We use a newly designed tumor-targeted oncolytic adenovirus (Ad-TD) to deliver non-secreting (ns) IL-12 to tumor cells and examine the therapeutic and toxic effects in Syrian hamster models of pancreatic cancer (PaCa). Strikingly, intraperitoneal delivery of Ad-TD-nsIL-12 significantly enhanced survival of animals with orthotopic PaCa and cured peritoneally disseminated PaCa with no toxic side effects, in contrast to the treatment with Ad-TD expressing unmodified IL-12. These findings offer renewed hope for development of IL-12-based treatments for cancer. Interleukin-12 (IL-12) is a potent immunotherapeutic agent.
Collapse
|
869
|
Yokoda R, Nagalo BM, Vernon B, Oklu R, Albadawi H, DeLeon TT, Zhou Y, Egan JB, Duda DG, Borad MJ. Oncolytic virus delivery: from nano-pharmacodynamics to enhanced oncolytic effect. Oncolytic Virother 2017; 6:39-49. [PMID: 29184854 PMCID: PMC5687448 DOI: 10.2147/ov.s145262] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
With the advancement of a growing number of oncolytic viruses (OVs) to clinical development, drug delivery is becoming an important barrier to overcome for optimal therapeutic benefits. Host immunity, tumor microenvironment and abnormal vascularity contribute to inefficient vector delivery. A number of novel approaches for enhanced OV delivery are under evaluation, including use of nanoparticles, immunomodulatory agents and complex viral–particle ligands along with manipulations of the tumor microenvironment. This field of OV delivery has quickly evolved to bioengineering of complex nanoparticles that could be deposited within the tumor using minimal invasive image-guided delivery. Some of the strategies include ultrasound (US)-mediated cavitation-enhanced extravasation, magnetic viral complexes delivery, image-guided infusions with focused US and targeting photodynamic virotherapy. In addition, strategies that modulate tumor microenvironment to decrease extracellular matrix deposition and increase viral propagation are being used to improve tumor penetration by OVs. Some involve modification of the viral genome to enhance their tumoral penetration potential. Here, we highlight the barriers to oncolytic viral delivery, and discuss the challenges to improving it and the perspectives of establishing new modes of active delivery to achieve enhanced oncolytic effects.
Collapse
Affiliation(s)
- Raquel Yokoda
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Bolni M Nagalo
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Brent Vernon
- Department of Biomedical Engineering, Arizona State University, Tempe
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Thomas T DeLeon
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Yumei Zhou
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Jan B Egan
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Dan G Duda
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Mitesh J Borad
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| |
Collapse
|
870
|
Sayour EJ, Mitchell DA. Immunotherapy for Pediatric Brain Tumors. Brain Sci 2017; 7:brainsci7100137. [PMID: 29065490 PMCID: PMC5664064 DOI: 10.3390/brainsci7100137] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/20/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Malignant brain tumors are the most common cause of solid cancer death in children. New targeted therapies are vital to improve treatment outcomes, but must be developed to enable trafficking across the blood brain barrier (BBB). Since activated T cells cross the BBB, cancer immunotherapy can be harnessed to unlock the cytotoxic potential of the immune system. However, standard of care treatments (i.e., chemotherapy and radiation) applied concomitant to pediatric brain tumor immunotherapy may abrogate induction of immunotherapeutic responses. This review will discuss the development of immunotherapies within this paradigm using emerging approaches being investigated in phase I/II trials in children with refractory brain tumors, including checkpoint inhibitors, vaccine immunotherapy, and adoptive cell therapy.
Collapse
Affiliation(s)
- Elias J Sayour
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| | - Duane A Mitchell
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
871
|
Santry LA, McAusland TM, Susta L, Wood GA, Major PP, Petrik JJ, Bridle BW, Wootton SK. Production and Purification of High-Titer Newcastle Disease Virus for Use in Preclinical Mouse Models of Cancer. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 9:181-191. [PMID: 29556508 PMCID: PMC5854916 DOI: 10.1016/j.omtm.2017.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/12/2017] [Indexed: 12/12/2022]
Abstract
Newcastle disease virus (NDV) is a single-stranded, negative-sense RNA virus in the Paramyxoviridae family. Although primarily an avian pathogen, NDV is a potent oncolytic virus that has been shown to be safe and effective in a variety of preclinical cancer models and human clinical trials. To produce virus for oncolytic trials, NDV is commonly amplified in embryonated chicken eggs and purified from the allantoic fluid. Conventional methods for purifying virus from allantoic fluid often result in relatively low-titer preparations containing high levels of impurities, including immunogenic chicken host cell proteins from allantoic fluid. However, large quantities of virus need to be delivered intravenously to administer oncolytic NDV systemically to mice. This route of administration requires virus preparations that are both highly concentrated (to enable delivery of small volumes) and highly pure (to limit toxic effects from contaminants). Given the accumulation of promising preclinical and clinical data demonstrating the efficacy of NDV as an oncolytic agent, strategies for increasing the titer and purity of NDV preparations are sorely needed to allow for effective intravenous administration in mice. Here, we describe an optimized protocol for the rescue, production, and purification of high-titer in vivo-grade NDV for preclinical studies in mouse models.
Collapse
Affiliation(s)
- Lisa A Santry
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Thomas M McAusland
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Pierre P Major
- Juravinski Cancer Centre, 699 Concession Street, Hamilton, ON L8V 5C2, Canada
| | - Jim J Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Byram W Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
872
|
Abstract
Purpose of Review This article provides a brief overview of the role that infections play in cancer emergence and cancer treatment. Recent Findings A select number of pathogens have been reported to increase the incidence of specific cancers (directly through altering gene expression or indirectly through inducing chronic inflammation). These have been referred to as oncogenic pathogens. Conversely, a subset of pathogens has been demonstrated to preferentially cause lysis of tumor cells, leading to tumor regression and improved anti-tumor immunity. These have been termed oncolytic pathogens. However, the contribution of non-oncogenic, non-oncolytic pathogens to both tumor growth and regression is likewise being increasingly recognized. Summary Pathogens have both the ability to cause and cure cancer. However, the mechanisms underlying these pathogen-mediated outcomes are not fully understood. With the recent emergence of interest in the immunotherapy of cancer, it is important that future studies focus specifically on preventing the negative effects of oncogenic infections, deconstructing the positive role of oncolytic pathogens, and finally providing insight into the dual roles of non-oncolytic, non-oncogenic pathogens so that anti-pathogen immune responses can be harnessed as a transformative means to treat cancer.
Collapse
|
873
|
Insertion of the Type-I IFN Decoy Receptor B18R in a miRNA-Tagged Semliki Forest Virus Improves Oncolytic Capacity but Results in Neurotoxicity. MOLECULAR THERAPY-ONCOLYTICS 2017; 7:67-75. [PMID: 29159280 PMCID: PMC5684435 DOI: 10.1016/j.omto.2017.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/02/2017] [Indexed: 12/28/2022]
Abstract
Oncolytic Semliki Forest virus (SFV) has been suggested as a potential candidate for the treatment of glioblastoma and neuroblastoma. However, the oncolytic capacity of SFV is restricted by the anti-viral type-I interferon (IFN) response. The aim of this study was to increase the oncolytic capacity of a microRNA target tagged SFV against glioblastoma by arming it with the Vaccinia-virus-encoded type-I IFN decoy receptor B18R (SFV4B18RmiRT) to neutralize type-I IFN response. Expression of B18R by SFV4B18RmiRT aided neutralization of IFN-β, which was shown by reduced STAT-1 phosphorylation and improved virus spread in plaque assays. B18R expression by SFV4 increased its oncolytic capacity in vitro against murine glioblastoma (CT-2A), regardless of the presence of exogenous IFN-β. Both SFV4B18RmiRT and SFV4miRT treatments controlled tumor growth in mice with syngeneic orthotopic gliomablastoma (CT-2A). However, treatment with SFV4B18RmiRT induced severe neurological symptoms in some mice because of virus replication in the healthy brain. Neither neurotoxicity nor virus replication in the brain was observed when SFV4miRT was administered. In summary, our results indicate that the oncolytic capacity of SFV4 was improved in vitro and in vivo by incorporation of B18R, but neurotoxicity of the virus was increased, possibly due to loss of microRNA targets.
Collapse
|
874
|
Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv Drug Deliv Rev 2017; 120:89-107. [PMID: 28736304 DOI: 10.1016/j.addr.2017.07.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 12/18/2022]
Abstract
Cancer progression to metastatic stages is still unmanageable and the promise of effective anti-metastatic therapy remains largely unmet, emphasizing the need to develop novel therapeutics. The special focus here is on cancer stem cells (CSC) as the seed of tumor initiation, epithelial-mesenchymal transition, chemoresistance and, as a consequence, drivers of metastatic dissemination. We report on targeted therapies gearing towards the CSC's internal and membrane-anchored markers using agents such as antibody derivatives, nucleic therapeutics, small molecules and genetic payloads. Another emphasis lies on novel proceedings envisaged to deliver current and prospective therapies to the target sites using newest viral and non-viral vector technologies. In this review, we summarize recent progress and remaining challenges in therapeutic strategies to combat CSC.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany.
| | - Manish Solanki
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| | - Ottmar Herchenröder
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| |
Collapse
|
875
|
Zhao X, Ouyang W, Chester C, Long S, Wang N, He Z. Cytokine-induced killer cell delivery enhances the antitumor activity of oncolytic reovirus. PLoS One 2017; 12:e0184816. [PMID: 28922411 PMCID: PMC5602626 DOI: 10.1371/journal.pone.0184816] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 08/31/2017] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OV) have recently emerged as a promising therapeutic modality in cancer treatment. OV selectively infect and kill tumor cells, while sparing untransformed cells. The direct cytotoxic effects combined with the capacity to trigger an immune response make OV an appealing combination partner in the burgeoning field of cancer immunotherapy. One of the leading OV therapeutic candidates is the double-stranded RNA virus reovirus. In order to improve the oncolytic activity of reovirus and allow for systemic administration despite the prevalence of neutralizing antibodies, cytokine-induced killer (CIK) cells were explored as cell carriers for reovirus delivery. In this study, CIK cells were successfully loaded with reovirus ex vivo, and viral replication was limited in CIK cells. Confocal microscopy and flow cytometry demonstrated that CIK cells retained reovirus on the surface. Moreover, CIK cells could promote reovirus infection of tumor cells in the presence of neutralizing antibodies; meanwhile, cytotoxicity of CIK cells was increased after loading with reovirus. These findings support further investigation of reovirus and CIK combination for antitumor therapy.
Collapse
Affiliation(s)
- Xing Zhao
- Stem Cell and Tissue Engineering Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Weiwei Ouyang
- Department of Thoracic Oncology, Affiliated Hospital of Guizhou Medical University, and Guizhou Cancer Hospital, Guiyang, Guizhou, China
| | - Cariad Chester
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California, United States of America
| | - Shiqi Long
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Nianxue Wang
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhixu He
- Stem Cell and Tissue Engineering Research Center, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
876
|
Zhang J, Lai W, Li Q, Yu Y, Jin J, Guo W, Zhou X, Liu X, Wang Y. A novel oncolytic adenovirus targeting Wnt signaling effectively inhibits cancer-stem like cell growth via metastasis, apoptosis and autophagy in HCC models. Biochem Biophys Res Commun 2017; 491:469-477. [PMID: 28698142 DOI: 10.1016/j.bbrc.2017.07.041] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 07/08/2017] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs), which are highly differentiated and self-renewing, play an important role in the occurrence, therapeutic resistant and metastasis of hepatacellular carcinoma (HCC). Oncolytic adenoviruses have targeted killing effect on tumor cells, and are invoked as candidate drugs for cancer treatment. We designed a dual-regulated oncolytic adenovirus Ad.wnt-E1A(△24bp)-TSLC1 that targets Wnt and Rb signaling pathways respectively, and carries the tumor suppressor gene, TSLC1. Previous studies have demonstrated that oncolytic adenovirus mediated TSLC1can target liver cancer and exhibit significant cytotoxicity. However, whether Ad.wnt-E1A(△24bp)-TSLC1 can effectively eliminate liver CSCs remains to be explored. We first used the spheroid culture to enrich the liver CSCs-like cells, and detected the self-renewal capacity, differentiation, drug resistance and tumorigenicity. The results showed that Ad-wnt-E1A(△24bp)-TSLC1 could effectively lead to autophagic death. In addition, recombinant adenovirus effectively induced the apoptosis, inhibit metastasis of hepatic CSCs-like cells in vivo. Further animal experiments indicated that Ad-wnt-E1A(△24bp)-TSLC1could effectively inhibit the growth of transplanted tumor of hepatic CSCs and prolong the survival time of mice. Therefore, the novel oncolytic adenovirus Ad.wnt-E1A(△24bp)-TSLC1 has potential application as a therapeutic target for HCC stem cells.
Collapse
Affiliation(s)
- Jian Zhang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Weijie Lai
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Qiang Li
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Yang Yu
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Jin Jin
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Wan Guo
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Xiumei Zhou
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| | - Xinyuan Liu
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China.
| | - Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| |
Collapse
|
877
|
Jhawar SR, Thandoni A, Bommareddy PK, Hassan S, Kohlhapp FJ, Goyal S, Schenkel JM, Silk AW, Zloza A. Oncolytic Viruses-Natural and Genetically Engineered Cancer Immunotherapies. Front Oncol 2017; 7:202. [PMID: 28955655 PMCID: PMC5600978 DOI: 10.3389/fonc.2017.00202] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022] Open
Abstract
There has long been interest in innovating an approach by which tumor cells can be selectively and specifically targeted and destroyed. The discovery of viruses that lyse tumor cells, termed oncolytic viruses (OVs), has led to a revolution in the treatment of cancer. The potential of OVs to improve the therapeutic ratio is derived from their ability to preferentially infect and replicate in cancer cells while avoiding destruction of normal cells surrounding the tumor. Two main mechanisms exist through which these viruses are reported to improve outcomes: direct lysis of tumor cells and indirect augmentation of host anti-tumor immunity. With these factors in mind, viruses are chosen or modified to selectively target tumor cells, decrease pathogenicity to normal cells, decrease the antiviral immune response (to prevent viral clearance), and increase the antitumor immune response. While only one OV has been approved for the treatment of cancer in the United States, and only two other OVs have been approved worldwide, a wide spectrum of OVs are in various stages of preclinical development and in clinical trials. These viruses are being studied as alternatives and adjuncts to more traditional cancer therapies including surgical resection, chemotherapy, radiation, hormonal therapies, targeted therapies, and other immunotherapies. Here, we review the natural characteristics and genetically engineered modifications that enhance the effectiveness of OVs for the treatment of cancer.
Collapse
Affiliation(s)
- Sachin R Jhawar
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.,Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Robert Wood Johnson University Hospital, New Brunswick, NJ, United States
| | - Aditya Thandoni
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | | | - Suemair Hassan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | | | - Sharad Goyal
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.,Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Robert Wood Johnson University Hospital, New Brunswick, NJ, United States
| | - Jason M Schenkel
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Ann W Silk
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Andrew Zloza
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.,Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
878
|
Xin G, Schauder DM, Zander R, Cui W. Two is better than one: advances in pathogen-boosted immunotherapy and adoptive T-cell therapy. Immunotherapy 2017; 9:837-849. [PMID: 28877635 PMCID: PMC5941714 DOI: 10.2217/imt-2017-0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/11/2017] [Indexed: 01/31/2023] Open
Abstract
The recent tremendous successes in clinical trials take cancer immunotherapy into a new era and have attracted major attention from both academia and industry. Among the variety of immunotherapy strategies developed to boost patients' own immune systems to fight against malignant cells, the pathogen-based and adoptive cell transfer therapies have shown the most promise for treating multiple types of cancer. Pathogen-based therapies could either break the immune tolerance to enhance the effectiveness of cancer vaccines or directly infect and kill cancer cells. Adoptive cell transfer can induce a strong durable antitumor response, with recent advances including engineering dual specificity into T cells to recognize multiple antigens and improving the metabolic fitness of transferred cells. In this review, we focus on the recent prospects in these two areas and summarize some ongoing studies that represent potential advancements for anticancer immunotherapy, including testing combinations of these two strategies.
Collapse
Affiliation(s)
- Gang Xin
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
| | - David M Schauder
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
- Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Ryan Zander
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
| | - Weiguo Cui
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
- Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
879
|
Ceppi F, Beck-Popovic M, Bourquin JP, Renella R. Opportunities and challenges in the immunological therapy of pediatric malignancy: a concise snapshot. Eur J Pediatr 2017; 176:1163-1172. [PMID: 28803259 DOI: 10.1007/s00431-017-2982-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/24/2017] [Accepted: 08/02/2017] [Indexed: 12/18/2022]
Abstract
Over the last 50 years, collaborative clinical trials have reduced the number of children dying from pediatric cancer significantly. Unfortunately, certain tumor types have remained resistant to conventional surgical, radiotherapy and chemotherapy combinations, and relapsing and/or refractory disease remains associated with dismal outcomes. Recently, renewed attention has been given to the role for immunotherapies in pediatric oncology. In fact, these combine several attractive features, including (but possibly not limited to) the specificity for cancer cells, potentially in vivo persistence and longevity, and potency against refractory disease. In this narrative review designed for the academic pediatrician, we will concisely review the biological underpinnings behind the immunological therapy of pediatric neoplasms and illustrate the current humoral, cellular approaches, and novel drugs targeting the immune checkpoint, oncolytic viruses, and tumor vaccines. We will also comment on the future directions, challenges, and open questions faced by the field. What is Known: • Cancer immunotherapy drives immune cells and its humoral weaponry to eliminate tumor cells. • This occurs by recognizing antigens ideally expressed only on tumoral, but not normal/healthy, cells. What is New: • Clinical immunotherapy trials have shown responses in children with relapsing/refractory neoplasms. • Novel humoral/cellular immunotherapies, immune checkpoint inhibitors, oncolytic viruses, and tumor vaccines are currently being investigated in pediatric oncology.
Collapse
Affiliation(s)
- Francesco Ceppi
- Pediatric Hematology-Oncology Research Laboratory & Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, University Hospital of Lausanne, Lausanne, Switzerland
| | - Maja Beck-Popovic
- Pediatric Hematology-Oncology Research Laboratory & Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, University Hospital of Lausanne, Lausanne, Switzerland
| | - Jean-Pierre Bourquin
- Leukemia Research Program and Division of Pediatric Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Raffaele Renella
- Pediatric Hematology-Oncology Research Laboratory & Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, University Hospital of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
880
|
Deines P, Lachnit T, Bosch TCG. Competing forces maintain theHydrametaorganism. Immunol Rev 2017; 279:123-136. [DOI: 10.1111/imr.12564] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Peter Deines
- Zoological Institute; Christian Albrechts University Kiel; Kiel Germany
| | - Tim Lachnit
- Zoological Institute; Christian Albrechts University Kiel; Kiel Germany
| | | |
Collapse
|
881
|
Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 2017; 6:e1371896. [PMID: 29209572 PMCID: PMC5706611 DOI: 10.1080/2162402x.2017.1371896] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The goal of cancer immunotherapy is to establish new or boost pre-existing anticancer immune responses that eradicate malignant cells while generating immunological memory to prevent disease relapse. Over the past few years, immunomodulatory monoclonal antibodies (mAbs) that block co-inhibitory receptors on immune effectors cells - such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) - or their ligands - such as CD274 (best known as PD-L1) - have proven very successful in this sense. As a consequence, many of such immune checkpoint blockers (ICBs) have already entered the clinical practice for various oncological indications. Considerable attention is currently being attracted by a second group of immunomodulatory mAbs, which are conceived to activate co-stimulatory receptors on immune effector cells. Here, we discuss the mechanisms of action of these immunostimulatory mAbs and summarize recent progress in their preclinical and clinical development.
Collapse
Affiliation(s)
- Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- DKFZ-Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
882
|
Pease DF, Kratzke RA. Oncolytic Viral Therapy for Mesothelioma. Front Oncol 2017; 7:179. [PMID: 28884088 PMCID: PMC5573749 DOI: 10.3389/fonc.2017.00179] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/04/2017] [Indexed: 12/22/2022] Open
Abstract
The limited effectiveness of conventional therapy for malignant pleural mesothelioma demands innovative approaches to this difficult disease. Even with aggressive multimodality treatment of surgery, radiation, and/or chemotherapy, the median survival is only 1–2 years depending on stage and histology. Oncolytic viral therapy has emerged in the last several decades as a rapidly advancing field of immunotherapy studied in a wide spectrum of malignancies. Mesothelioma makes an ideal candidate for studying oncolysis given the frequently localized pattern of growth and pleural location providing access to direct intratumoral injection of virus. Therefore, despite being a relatively uncommon disease, the multitude of viral studies for mesothelioma can provide insight for applying such therapy to other malignancies. This article will begin with a review of the general principles of oncolytic therapy focusing on antitumor efficacy, tumor selectivity, and immune system activation. The second half of this review will detail results of preclinical models and human studies for oncolytic virotherapy in mesothelioma.
Collapse
Affiliation(s)
- Daniel F Pease
- Hematology-Oncology-Transplant, University of Minnesota, Minneapolis, MN, United States
| | - Robert A Kratzke
- Hematology-Oncology-Transplant, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
883
|
Guo ZS, Bartlett DL. Editorial of the Special Issue: Oncolytic Viruses as a Novel Form of Immunotherapy for Cancer. Biomedicines 2017; 5:biomedicines5030052. [PMID: 28837095 PMCID: PMC5618310 DOI: 10.3390/biomedicines5030052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 08/22/2017] [Accepted: 08/22/2017] [Indexed: 01/05/2023] Open
Abstract
Oncolytic viruses (OVs), either occurring naturally or through genetic engineering, can selectively infect, replicate in, and kill cancer cells, while leaving normal cells (almost) unharmed [...].
Collapse
Affiliation(s)
- Zong Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| |
Collapse
|
884
|
Affiliation(s)
- Mark D. Vincent
- The University of Western Ontario − Oncology; Ontario; Canada
| |
Collapse
|
885
|
Nounamo B, Liem J, Cannon M, Liu J. Myxoma Virus Optimizes Cisplatin for the Treatment of Ovarian Cancer In Vitro and in a Syngeneic Murine Dissemination Model. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:90-99. [PMID: 28875159 PMCID: PMC5573804 DOI: 10.1016/j.omto.2017.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/06/2017] [Indexed: 12/15/2022]
Abstract
A therapeutic approach to improve treatment outcome of ovarian cancer (OC) in patients is urgently needed. Myxoma virus (MYXV) is a candidate oncolytic virus that infects to eliminate OC cells. We found that in vitro MYXV treatment enhances cisplatin or gemcitabine treatment by allowing lower doses than the corresponding IC50 calculated for primary OC cells. MYXV also affected OC patient ascites-associated CD14+ myeloid cells, one of the most abundant immunological components of the OC tumor environment; without causing cell death, MYXV infection reduces the ability of these cells to secrete cytokines such as IL-10 that are signatures of the immunosuppressive tumor environment. We found that pretreatment with replication-competent but not replication-defective MYXV-sensitized tumor cells to later cisplatin treatments to drastically improve survival in a murine syngeneic OC dissemination model. We thus conclude that infection with replication-competent MYXV before cisplatin treatment markedly enhances the therapeutic benefit of chemotherapy. Treatment with replication-competent MYXV followed by cisplatin potentiated splenocyte activation and IFNγ expression, possibly by T cells, when splenocytes from treated mice were stimulated with tumor cell antigen ex vivo. The impact on immune responses in the tumor environment may thus contribute to the enhanced antitumor activity of combinatorial MYXV-cisplatin treatment.
Collapse
Affiliation(s)
- Bernice Nounamo
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA
| | - Jason Liem
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA
| | - Martin Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205-7199, USA.,The Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
886
|
Le Boeuf F, Gebremeskel S, McMullen N, He H, Greenshields AL, Hoskin DW, Bell JC, Johnston B, Pan C, Duncan R. Reovirus FAST Protein Enhances Vesicular Stomatitis Virus Oncolytic Virotherapy in Primary and Metastatic Tumor Models. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:80-89. [PMID: 28856238 PMCID: PMC5562180 DOI: 10.1016/j.omto.2017.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/01/2017] [Indexed: 12/28/2022]
Abstract
The reovirus fusion-associated small transmembrane (FAST) proteins are the smallest known viral fusogens (∼100–150 amino acids) and efficiently induce cell-cell fusion and syncytium formation in multiple cell types. Syncytium formation enhances cell-cell virus transmission and may also induce immunogenic cell death, a form of apoptosis that stimulates immune recognition of tumor cells. These properties suggest that FAST proteins might serve to enhance oncolytic virotherapy. The oncolytic activity of recombinant VSVΔM51 (an interferon-sensitive vesicular stomatitis virus [VSV] mutant) encoding the p14 FAST protein (VSV-p14) was compared with a similar construct encoding GFP (VSV-GFP) in cell culture and syngeneic BALB/c tumor models. Compared with VSV-GFP, VSV-p14 exhibited increased oncolytic activity against MCF-7 and 4T1 breast cancer spheroids in culture and reduced primary 4T1 breast tumor growth in vivo. VSV-p14 prolonged survival in both primary and metastatic 4T1 breast cancer models, and in a CT26 metastatic colon cancer model. As with VSV-GFP, VSV-p14 preferentially replicated in vivo in tumors and was cleared rapidly from other sites. Furthermore, VSV-p14 increased the numbers of activated splenic CD4, CD8, natural killer (NK), and natural killer T (NKT) cells, and increased the number of activated CD4 and CD8 cells in tumors. FAST proteins may therefore provide a multi-pronged approach to improving oncolytic virotherapy via syncytium formation and enhanced immune stimulation.
Collapse
Affiliation(s)
- Fabrice Le Boeuf
- 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
| | - Simon Gebremeskel
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Nichole McMullen
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Han He
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | | | - David W Hoskin
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - John C Bell
- 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
| | - Brent Johnston
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada.,Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada.,Department of Pediatrics, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Chungen Pan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Roy Duncan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada.,Department of Pediatrics, Dalhousie University, Halifax, NS B3H4R2, Canada.,Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H4R2, Canada
| |
Collapse
|
887
|
Boisgerault N, Grégoire M, Fonteneau JF. Viral cancer therapies: are they ready for combination with other immunotherapies? Future Oncol 2017; 13:1569-1571. [PMID: 28776392 DOI: 10.2217/fon-2017-0135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
| | - Marc Grégoire
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | | |
Collapse
|
888
|
Svyatchenko VA, Ternovoy VA, Kiselev NN, Demina AV, Loktev VB, Netesov SV, Chumakov PM. Bioselection of coxsackievirus B6 strain variants with altered tropism to human cancer cell lines. Arch Virol 2017; 162:3355-3362. [PMID: 28766058 DOI: 10.1007/s00705-017-3492-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/02/2017] [Indexed: 12/21/2022]
Abstract
Cancer cells develop increased sensitivity to members of many virus families and, in particular, can be efficiently infected and lysed by many low-pathogenic human enteroviruses. However, because of their great genetic heterogeneity, cancer cells display different levels of sensitivity to particular enterovirus strains, which may substantially limit the chances of a positive clinical response. We show that a non-pathogenic strain of coxsackievirus B6 (LEV15) can efficiently replicate to high titers in the malignant human cell lines C33A, DU145, AsPC-1 and SK-Mel28, although it displays much lower replication efficiency in A431 and A549 cells and very limited replication ability in RD and MCF7 cells, as well as in the normal lung fibroblast cell line MRC-5 and the immortalized mammary epithelial cell line MCF10A. By serial passaging in RD, MCF7 and A431 cells, we obtained LEV15 strain variants that had acquired high replication capacity in the appropriate carcinoma cell lines without losing their high replication capability in the original set of cancer cell lines and had limited replication capability in untransformed cells. The strains demonstrated improved oncolytic properties in nude-mouse xenografts. We identified nucleotide changes responsible for the phenotypes and suggest a bioselection approach for a generation of oncolytic virus strains with a wider spectrum of affected tumors.
Collapse
Affiliation(s)
- Victor A Svyatchenko
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk, Russia
| | - Vladimir A Ternovoy
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk, Russia
| | - Nikolai N Kiselev
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk, Russia
| | - Anna V Demina
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk, Russia
| | - Valery B Loktev
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Sergey V Netesov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Peter M Chumakov
- Novosibirsk State University, Novosibirsk, Russia.
- Engelhardt Institute of Molecular Biology, Moscow, Russia.
- M.P. Chumakov Institute of Poliomyelitis and Viral Encephalitides, Federal Scientific Center on Research and Development of Immunobiology Products, Moscow, Russia.
| |
Collapse
|
889
|
Masemann D, Boergeling Y, Ludwig S. Employing RNA viruses to fight cancer: novel insights into oncolytic virotherapy. Biol Chem 2017; 398:891-909. [DOI: 10.1515/hsz-2017-0103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/08/2017] [Indexed: 12/13/2022]
Abstract
Abstract
Within recent decades, viruses that specifically target tumor cells have emerged as novel therapeutic agents against cancer. These viruses do not only act via their cell-lytic properties, but also harbor immunostimulatory features to re-direct the tumor microenvironment and stimulate tumor-directed immune responses. Furthermore, oncolytic viruses are considered to be superior to classical cancer therapies due to higher selectivity towards tumor cell destruction and, consequently, less collateral damage of non-transformed healthy tissue. In particular, the field of oncolytic RNA viruses is rapidly developing since these agents possess alternative tumor-targeting strategies compared to established oncolytic DNA viruses. Thus, oncolytic RNA viruses have broadened the field of virotherapy facilitating new strategies to fight cancer. In addition to several naturally occurring oncolytic viruses, genetically modified RNA viruses that are armed to express foreign factors such as immunostimulatory molecules have been successfully tested in early clinical trials showing promising efficacy. This review aims to provide an overview of the most promising RNA viruses in clinical development, to summarize the current knowledge of clinical trials using these viral agents, and to discuss the main issues as well as future perspectives of clinical approaches using oncolytic RNA viruses.
Collapse
|
890
|
O’Cathail SM, Pokrovska TD, Maughan TS, Fisher KD, Seymour LW, Hawkins MA. Combining Oncolytic Adenovirus with Radiation-A Paradigm for the Future of Radiosensitization. Front Oncol 2017; 7:153. [PMID: 28791251 PMCID: PMC5523729 DOI: 10.3389/fonc.2017.00153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/28/2017] [Indexed: 01/03/2023] Open
Abstract
Oncolytic viruses and radiotherapy represent two diverse areas of cancer therapy, utilizing quite different treatment modalities and with non-overlapping cytotoxicity profiles. It is, therefore, an intriguing possibility to consider that oncolytic ("cancer-killing") viruses may act as cancer-selective radiosensitizers, enhancing the therapeutic consequences of radiation treatment on tumors while exerting minimal effects on normal tissue. There is a solid mechanistic basis for this potential synergy, with many viruses having developed strategies to inhibit cellular DNA repair pathways in order to protect themselves, during genome replication, from unwanted interference by cell processes that are normally triggered by DNA damage. Exploiting these abilities to inhibit cellular DNA repair following damage by therapeutic irradiation may well augment the anticancer potency of the approach. In this review, we focus on oncolytic adenovirus, the most widely developed and best understood oncolytic virus, and explore its various mechanisms for modulating cellular DNA repair pathways. The most obvious effects of the various adenovirus serotypes are to interfere with activity of the MRE11-Rad50-Nbs1 complex, temporally one of the first sensors of double-stranded DNA damage, and inhibition of DNA ligase IV, a central repair enzyme for healing double-stranded breaks by non-homologous end joining (NHEJ). There have been several preclinical and clinical studies of this approach and we assess the current state of progress. In addition, oncolytic viruses provide the option to promote a localized proinflammatory response, both by mediating immunogenic death of cancer cells by oncosis and also by encoding and expressing proinflammatory biologics within the tumor microenvironment. Both of these approaches provide exciting potential to augment the known immunological consequences of radiotherapy, aiming to develop systems capable of creating a systemic anticancer immune response following localized tumor treatment.
Collapse
Affiliation(s)
- Sean M. O’Cathail
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Timothy S. Maughan
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Kerry D. Fisher
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Maria A. Hawkins
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
891
|
Turbocharging vaccines: emerging adjuvants for dendritic cell based therapeutic cancer vaccines. Curr Opin Immunol 2017; 47:35-43. [PMID: 28732279 DOI: 10.1016/j.coi.2017.06.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/22/2017] [Indexed: 12/19/2022]
Abstract
Development of therapeutic cancer vaccines has been hindered by the many pro-tumorigenic mechanisms at play in cancer patients that serve to suppress both antigen presenting cells and T cells. In face of these obstacles, cancer vaccines are most likely to promote anti-tumorigenic immune responses only when formulated with strong adjuvants, and in combination with new immune interventions designed to reverse immune suppression and exhaustion of T cells in the tumor microenvironment. Dendritic cells (DCs) are often termed 'nature's adjuvant' due to their exceptional capacity for initiating both innate and adaptive immune responses. Hence, the past decade has witnessed a flurry of activity in testing DC based immunotherapies for cancer intervention. In this review we will discuss advances in conventional adjuvants and provide insight into new adjuvants as they pertain to DC cancer therapy.
Collapse
|
892
|
The development of activatable lytic peptides for targeting triple negative breast cancer. Cell Death Discov 2017; 3:17037. [PMID: 29263848 PMCID: PMC5629628 DOI: 10.1038/cddiscovery.2017.37] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/14/2017] [Accepted: 05/22/2017] [Indexed: 12/28/2022] Open
Abstract
Cytolytic peptides are an emerging class of promising cancer therapeutics shown to overcome drug resistance. They eliminate cancer cells via disruption of the phospholipid bilayer of cell membranes, a mechanism that differentiates it from traditional treatments. However, applications of lytic peptides via systematic administration are hampered by nonspecific toxicity. Here, we describe activatable, masked lytic peptides that are conjugated with anionic peptides via a cleavable linker sensitive to matrix metalloproteinases (Ac-w-βA-e8-XPLG*LAG-klUklUkklUklUk-NH2; lower case letters in the sequences represent D-amino-acids, U=Aib, α-aminoisobutyric acid, *cleavage site). The peptides were activated upon being introduced into the triple negative breast cancer cell line MDA-MB-231, which overexpresses secreted matrix metalloproteinases, to selectively cleave the peptide linker. Our results indicate that the activatable design could be applied to improve the targeting ability of lytic peptides.
Collapse
|
893
|
Eissa IR, Naoe Y, Bustos-Villalobos I, Ichinose T, Tanaka M, Zhiwen W, Mukoyama N, Morimoto T, Miyajima N, Hitoki H, Sumigama S, Aleksic B, Kodera Y, Kasuya H. Genomic Signature of the Natural Oncolytic Herpes Simplex Virus HF10 and Its Therapeutic Role in Preclinical and Clinical Trials. Front Oncol 2017; 7:149. [PMID: 28770166 PMCID: PMC5509757 DOI: 10.3389/fonc.2017.00149] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/26/2017] [Indexed: 12/19/2022] Open
Abstract
Oncolytic viruses (OVs) are opening new possibilities in cancer therapy with their unique mechanism of selective replication within tumor cells and triggering of antitumor immune responses. HF10 is an oncolytic herpes simplex virus-1 with a unique genomic structure that has non-engineered deletions and insertions accompanied by frame-shift mutations, in contrast to the majority of engineered OVs. At the genetic level, HF10 naturally lacks the expression of UL43, UL49.5, UL55, UL56, and latency-associated transcripts, and overexpresses UL53 and UL54. In preclinical studies, HF10 replicated efficiently within tumor cells with extensive cytolytic effects and induced increased numbers of activated CD4+ and CD8+ T cells and natural killer cells within the tumor, leading to a significant reduction in tumor growth and prolonged survival rates. Investigator-initiated clinical studies of HF10 have been completed in recurrent breast carcinoma, head and neck cancer, and unresectable pancreatic cancer in Japan. Phase I trials were subsequently completed in refractory superficial cancers and melanoma in the United States. HF10 has been demonstrated to have a high safety margin with low frequency of adverse effects in all treated patients. Interestingly, HF10 antigens were detected in pancreatic carcinoma over 300 days after treatment with infiltration of CD4+ and CD8+ T cells, which enhanced the immune response. To date, preliminary results from a Phase II trial have indicated that HF10 in combination with ipilimumab (anti-CTLA-4) is safe and well tolerated, with high antitumor efficacy. Improvement of the effect of ipilimumab was observed in patients with stage IIIb, IIIc, or IV unresectable or metastatic melanoma. This review provides a concise description of the genomic functional organization of HF10 compared with talimogene laherparepvec. Furthermore, this review focuses on HF10 in cancer treatment as monotherapy as well as in combination therapy through a concise description of all preclinical and clinical data. In addition, we will address approaches for future directions in HF10 studies as cancer therapy.
Collapse
Affiliation(s)
- Ibrahim Ragab Eissa
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Faculty of Science, Tanta University, Tanta, Egypt
| | - Yoshinori Naoe
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Itzel Bustos-Villalobos
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Toru Ichinose
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | | | - Wu Zhiwen
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Nobuaki Mukoyama
- Department of Otolaryngology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Taishi Morimoto
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Noriyuki Miyajima
- Department of Transplantation and Endocrine Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hasegawa Hitoki
- Office of International Affairs, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Seiji Sumigama
- Office of International Affairs, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Branko Aleksic
- Office of International Affairs, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hideki Kasuya
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
894
|
Guillard S, Kolasinska-Zwierz P, Debreczeni J, Breed J, Zhang J, Bery N, Marwood R, Tart J, Overman R, Stocki P, Mistry B, Phillips C, Rabbitts T, Jackson R, Minter R. Structural and functional characterization of a DARPin which inhibits Ras nucleotide exchange. Nat Commun 2017; 8:16111. [PMID: 28706291 PMCID: PMC5519984 DOI: 10.1038/ncomms16111] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/30/2017] [Indexed: 12/19/2022] Open
Abstract
Ras mutations are the oncogenic drivers of many human cancers and yet there are still no approved Ras-targeted cancer therapies. Inhibition of Ras nucleotide exchange is a promising new approach but better understanding of this mechanism of action is needed. Here we describe an antibody mimetic, DARPin K27, which inhibits nucleotide exchange of Ras. K27 binds preferentially to the inactive Ras GDP form with a Kd of 4 nM and structural studies support its selectivity for inactive Ras. Intracellular expression of K27 significantly reduces the amount of active Ras, inhibits downstream signalling, in particular the levels of phosphorylated ERK, and slows the growth in soft agar of HCT116 cells. K27 is a potent, non-covalent inhibitor of nucleotide exchange, showing consistent effects across different isoforms of Ras, including wild-type and oncogenic mutant forms. Ras is mutated in many cancers, but so far no drug targeting Ras is in clinical use despite great efforts. Here the authors structurally and functionally characterize a DARPin that potently inhibits the nucleotide exchange of Ras, which might facilitate the development of Ras-targeted therapies.
Collapse
Affiliation(s)
- Sandrine Guillard
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| | - Paulina Kolasinska-Zwierz
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| | - Judit Debreczeni
- Discovery Sciences, Innovative Medicines and Early Development, AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - Jason Breed
- Discovery Sciences, Innovative Medicines and Early Development, AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - Jing Zhang
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Nicolas Bery
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Rose Marwood
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| | - Jon Tart
- Discovery Sciences, Innovative Medicines and Early Development, AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - Ross Overman
- Discovery Sciences, Innovative Medicines and Early Development, AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - Pawel Stocki
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| | - Bina Mistry
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| | - Christopher Phillips
- Discovery Sciences, Innovative Medicines and Early Development, AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - Terence Rabbitts
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Ronald Jackson
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| | - Ralph Minter
- Antibody Discovery and Protein Engineering, MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, UK
| |
Collapse
|
895
|
Stem cell-released oncolytic herpes simplex virus has therapeutic efficacy in brain metastatic melanomas. Proc Natl Acad Sci U S A 2017; 114:E6157-E6165. [PMID: 28710334 DOI: 10.1073/pnas.1700363114] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The recent Food and Drug Administration approval of immunogenic oncolytic virus (OV) has opened a new era in the treatment of advanced melanoma; however, approximately 50% of patients with melanoma develop brain metastasis, and currently there are no beneficial treatment options for such patients. To model the progression of metastases seen in patients and to overcome the hurdles of systemic delivery of OV, we developed melanoma brain metastasis models in immunocompromised and immunocompetent mice, and tested the fate and efficacy of oncolytic herpes simplex virus (oHSV)-armed mesenchymal stem cells (MSCs). Using brain-seeking patient-derived melanoma cells and real-time in vivo imaging, we show a widespread distribution of micrometastases and macrometastases in the brain, recapitulating the progression of multifoci metastases seen in patients. We armed MSCs with different oHSV variants (MSC-oHSV) and found that intracarotid administration of MSC-oHSV, but not of purified oHSV alone, effectively tracks metastatic tumor lesions and significantly prolongs the survival of brain tumor-bearing mice. In a syngeneic model of melanoma brain metastasis, a combination of MSC-oHSV and PD-L1 blockade increases IFNγ-producing CD8+ tumor-infiltrating T lymphocytes and results in a profound extension of the median survival of treated animals. This study thus demonstrates the utility of MSCs as OV carriers to disseminated brain lesions, and provides a clinically applicable therapeutic platform to target melanoma brain metastasis.
Collapse
|
896
|
Oncolytic measles virus enhances antitumour responses of adoptive CD8 +NKG2D + cells in hepatocellular carcinoma treatment. Sci Rep 2017; 7:5170. [PMID: 28701757 PMCID: PMC5507973 DOI: 10.1038/s41598-017-05500-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023] Open
Abstract
There is an urgent need for novel effective treatment for hepatocellular carcinoma (HCC). Oncolytic viruses (OVs) not only directly lyse malignant cells, but also induce potent antitumour immune responses. The potency and precise mechanisms of antitumour immune activation by attenuated measles virus remain unclear. In this study, we investigated the potency of the measles virus vaccine strain Edmonston (MV-Edm) in improving adoptive CD8+NKG2D+ cells for HCC treatment. We show that MV-Edm-infected HCC enhanced the antitumour activity of CD8+NKG2D+ cells, mediated by at least three distinct mechanisms. First, MV-Edm infection compelled HCC cells to express the specific NKG2D ligands MICA/B, which may contribute to the activation of CD8+NKG2D+ cells. Second, MV-Edm-infected HCC cells stimulated CD8+NKG2D+ cells to express high level of FasL resulting in enhanced induction of apoptosis. Third, intratumoural administration of MV-Edm enhanced infiltration of intravenously injected CD8+NKG2D+ cells. Moreover, we found that MV-Edm and adoptive CD8+NKG2D+ cells, either administered alone or combined, upregulated the immune suppressive enzyme indoleamine 2,3-dioxygenase 1 (IDO1) in HCC. Elimination of IDO1 by fludarabine enhanced antitumour responses. Taken together, our data provide a novel and clinically relevant strategy for treatment of HCC.
Collapse
|
897
|
Abstract
From the application of Coley's toxin in the early 1900s to the present clinical trials using immune checkpoint regulatory inhibitors, the history of cancer immunotherapy has consisted of extremely high levels of enthusiasm after anecdotal case reports of enormous success, followed by decreasing levels of enthusiasm as the results of controlled clinical trials are available. In this review, this pattern will be documented for the various immunotherapeutic approaches over the years. The sole exception being vaccination against cancer causing viruses, which have already prevented thousands of cancers. We can only hope that the present high level of enthusiasm for the use of immune stimulation by removal of blocks to cancer immunity will be more productive than the incremental improvements using previous immunotherapies.
Collapse
Affiliation(s)
- Stewart Sell
- Wadsworth Center, New York State Department of Health and Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| |
Collapse
|
898
|
Miles LA, Burga LN, Gardner EE, Bostina M, Poirier JT, Rudin CM. Anthrax toxin receptor 1 is the cellular receptor for Seneca Valley virus. J Clin Invest 2017. [PMID: 28650343 DOI: 10.1172/jci93472] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Seneca Valley virus (SVV) is an oncolytic picornavirus with selective tropism for neuroendocrine cancers. It has shown promise as a cancer therapeutic in preclinical studies and early-phase clinical trials. Here, we have identified anthrax toxin receptor 1 (ANTXR1) as the receptor for SVV using genome-wide loss-of-function screens. ANTXR1 is necessary for permissivity in vitro and in vivo. However, robust SVV replication requires an additional innate immune defect. We found that SVV interacts directly and specifically with ANTXR1, that this interaction is required for SVV binding to permissive cells, and that ANTXR1 expression is necessary and sufficient for infection in cell lines with decreased expression of antiviral IFN genes at baseline. Finally, we identified the region of the SVV capsid that is responsible for receptor recognition using cryoelectron microscopy of the SVV-ANTXR1-Fc complex. These studies identify ANTXR1, a class of receptor that is shared by a mammalian virus and a bacterial toxin, as the cellular receptor for SVV.
Collapse
Affiliation(s)
- Linde A Miles
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Eric E Gardner
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mihnea Bostina
- Department of Microbiology and Immunology and.,Otago Centre for Electron Microscopy, University of Otago, Dunedin, New Zealand
| | - John T Poirier
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Charles M Rudin
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
899
|
Yin J, Markert JM, Leavenworth JW. Modulation of the Intratumoral Immune Landscape by Oncolytic Herpes Simplex Virus Virotherapy. Front Oncol 2017; 7:136. [PMID: 28695111 PMCID: PMC5483455 DOI: 10.3389/fonc.2017.00136] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 06/09/2017] [Indexed: 12/28/2022] Open
Abstract
Vaccines and immunotherapeutic approaches to cancers with the advent of immune checkpoint inhibitors and chimeric antigen receptor-modified T cells have recently demonstrated preclinical success and entered clinical trials. Despite advances in these approaches and combinatorial therapeutic regimens, depending on the nature of the cancer and the immune and metabolic landscape within the tumor microenvironment, current immunotherapeutic modalities remain inadequate. Recent clinical trials have demonstrated clear evidence of significant, and sometimes dramatic, antitumor activity, and long-term survival effects of a variety of oncolytic viruses (OVs), particularly oncolytic herpes simplex virus (oHSV). Acting as a multifaceted gene therapy vector and potential adjuvant-like regimens, oHSV can carry genes encoding immunostimulatory molecules in its genome. The oncolytic effect of oHSV and the inflammatory response that the virus stimulates provide a one-two punch at attacking tumors. However, mechanisms underlying oHSV-induced restoration of intratumoral immunosuppression demand extensive research in order to further improve its therapeutic efficacy. In this review, we discuss the current OV-based therapy, with a focus on the unique aspects of oHSV-initiated antiviral and antitumor immune responses, arising from virus-mediated immunological cell death to intratumoral innate and adaptive immunity.
Collapse
Affiliation(s)
- Jie Yin
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
900
|
Cockle JV, Brüning-Richardson A, Scott KJ, Thompson J, Kottke T, Morrison E, Ismail A, Carcaboso AM, Rose A, Selby P, Conner J, Picton S, Short S, Vile R, Melcher A, Ilett E. Oncolytic Herpes Simplex Virus Inhibits Pediatric Brain Tumor Migration and Invasion. Mol Ther Oncolytics 2017; 5:75-86. [PMID: 28547002 PMCID: PMC5435599 DOI: 10.1016/j.omto.2017.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 04/25/2017] [Indexed: 12/13/2022] Open
Abstract
Pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine glioma (DIPG) are invasive tumors with poor survival. Oncolytic virotherapy, initially devised as a direct cytotoxic treatment, is now also known to act via immune-mediated mechanisms. Here we investigate a previously unreported mechanism of action: the inhibition of migration and invasion in pediatric brain tumors. We evaluated the effect of oncolytic herpes simplex virus 1716 (HSV1716) on the migration and invasion of pHGG and DIPG both in vitro using 2D (scratch assay, live cell imaging) and 3D (spheroid invasion in collagen) assays and in vivo using an orthotopic xenograft model of DIPG invasion. HSV1716 inhibited migration and invasion in pHGG and DIPG cell lines. pHGG cells demonstrated reduced velocity and changed morphology in the presence of virus. HSV1716 altered pHGG cytoskeletal dynamics by stabilizing microtubules, inhibiting glycogen synthase kinase-3, and preventing localized clustering of adenomatous polyposis coli (APC) to the leading edge of cells. HSV1716 treatment also reduced tumor infiltration in a mouse orthotopic xenograft DIPG model. Our results demonstrate that HSV1716 targets the migration and invasion of pHGG and DIPG and indicates the potential of an oncolytic virus (OV) to be used as a novel anti-invasive treatment strategy for pediatric brain tumors.
Collapse
Affiliation(s)
- Julia V. Cockle
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
- Yorkshire Regional Centre for Paediatric Oncology and Haematology, Leeds General Infirmary, Leeds LS1 3EX, UK
| | | | - Karen J. Scott
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | - Jill Thompson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy Kottke
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ewan Morrison
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds LS9 7TF, UK
| | - Azam Ismail
- Department of Pathology, St. James’s University Hospital, Leeds LS9 7TF, UK
| | | | - Ailsa Rose
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | - Peter Selby
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | | | - Susan Picton
- Yorkshire Regional Centre for Paediatric Oncology and Haematology, Leeds General Infirmary, Leeds LS1 3EX, UK
| | - Susan Short
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | - Richard Vile
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Alan Melcher
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
- Institute of Cancer Research, London SM2 5NG, UK
| | - Elizabeth Ilett
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| |
Collapse
|