1
|
Saxena R, Gottlin EB, Campa MJ, He YW, Patz EF. Complement regulators as novel targets for anti-cancer therapy: A comprehensive review. Semin Immunol 2025; 77:101931. [PMID: 39826189 DOI: 10.1016/j.smim.2025.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/04/2025] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Cancer remains a formidable global health challenge requiring the continued exploration of innovative therapeutic approaches. While traditional treatment strategies including surgery, chemotherapy, and radiation therapy have had some success, primarily in early-stage disease, the quest for more targeted, personalized, safer, and effective therapies remains an ongoing pursuit. Over the past decade, significant advances in the field of tumor immunology have dramatically shifted a focus towards immunotherapy, although the ability to harness and coopt the immune system to treat cancer is still just beginning to be realized. One important area that has yet to be fully explored is the complement system, an integral part of innate immunity that has gathered attention recently as a source of potential targets for anti-cancer therapy. The complement system has a complex and context dependent role in cancer biology in that it not only contributes to immune surveillance but also may promote tumor progression. Complement regulators, including CD46, CD55, CD59, and complement factor H, exercise defined control over complement activation, and have also been acknowledged for their role in the tumor microenvironment. This review explores the intricate role of complement regulators in cancer development and progression, examining their potential as therapeutic targets, current strategies, challenges, and the evolving landscape of clinical research.
Collapse
Affiliation(s)
- Ruchi Saxena
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Elizabeth B Gottlin
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael J Campa
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Edward F Patz
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Wang Y, Cheng P. Arming oncolytic viruses with bispecific T cell engagers: The evolution and current status. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166962. [PMID: 37984801 DOI: 10.1016/j.bbadis.2023.166962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/22/2023]
Abstract
Oncolytic viruses (OVs) are emerging as therapeutically relevant anticancer agents as contemporary immunotherapy gains traction. Furthermore, OVs are an ideal platform for genetic modification to express therapeutic transgenes. Bispecific T cell engagers (BiTEs) can redirect T cells to tumor cells, resulting in targeted cytotoxicity. BiTEs have demonstrated success in hematological cancers but are rarely used in solid tumors. The drawbacks of BiTEs, including inadequate delivery and on-target-off-tumor activity have limited their efficacy. Combining OVs with BiTEs is a prospective area to investigate. This combined strategy can benefit from the best qualities of both therapies while overcoming the limitations.
Collapse
Affiliation(s)
- Yunmeng Wang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, PR China
| | - Ping Cheng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Effantin G, Hograindleur MA, Fenel D, Fender P, Vassal-Stermann E. Toward the understanding of DSG2 and CD46 interaction with HAdV-11 fiber, a super-complex analysis. J Virol 2023; 97:e0091023. [PMID: 37921471 PMCID: PMC10688334 DOI: 10.1128/jvi.00910-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 11/04/2023] Open
Abstract
IMPORTANCE The main limitation of oncolytic vectors is neutralization by blood components, which prevents intratumoral administration to patients. Enadenotucirev, a chimeric HAdV-11p/HAdV-3 adenovirus identified by bio-selection, is a low seroprevalence vector active against a broad range of human carcinoma cell lines. At this stage, there's still some uncertainty about tropism and primary receptor utilization by HAdV-11. However, this information is very important, as it has a direct influence on the effectiveness of HAdV-11-based vectors. The aim of this work is to determine which of the two receptors, DSG2 and CD46, is involved in the attachment of the virus to the host, and what role they play in the early stages of infection.
Collapse
Affiliation(s)
| | | | - Daphna Fenel
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Pascal Fender
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | |
Collapse
|
4
|
Letafati A, Ardekani OS, Naderisemiromi M, Fazeli MM, Jemezghani NA, Yavarian J. Oncolytic viruses against cancer, promising or delusion? Med Oncol 2023; 40:246. [PMID: 37458862 DOI: 10.1007/s12032-023-02106-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023]
Abstract
Cancer treatment is one of the most challenging topics in medical sciences. Different methods such as chemotherapy, tumor surgery, and immune checkpoint inhibitors therapy (ICIs) are potential approaches to treating cancer and killing tumor cells, but clinical studies have shown that they have been successful for a limited group of patients. Using viruses as a treatment can be considered as an effective treatment in the field of medicine. This is considered as a potential treatment, especially in comparison to chemotherapy, which has severe side effects related to the immune system. Most oncolytic viruses (OVs) have the potential to multiply in cancer cells, which are more than normal cells in malignant tissue and can induce immune responses. Therefore, tons of efforts and research have been started on the utilization of OVs as a treatment for cancer and have shown promising in treating cancers with less side effects. In this article, we have gathered studies about oncolytic viruses and their effectiveness in cancer treatment.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Author 1 Given name: [Omid Salahi] Last name [Ardekani], Author 2 Given name: [Mohammad Mehdi] Last name [Fazeli], Author 3 Given name: [Nillofar Asadi] Last name [Jemezghani]. Also, kindly confirm the details in the metadata are correct.Confirmed.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mina Naderisemiromi
- Department of Immunology, Faculty of Medicine and Health, The University of Manchester, Manchester, UK
| | - Mohammad Mehdi Fazeli
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | | - Jila Yavarian
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Gryciuk A, Rogalska M, Baran J, Kuryk L, Staniszewska M. Oncolytic Adenoviruses Armed with Co-Stimulatory Molecules for Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071947. [PMID: 37046608 PMCID: PMC10093006 DOI: 10.3390/cancers15071947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
In clinical trials, adenovirus vectors (AdVs) are commonly used platforms for human gene delivery therapy. High genome capacity and flexibility in gene organization make HAdVs suitable for cloning. Recent advancements in molecular techniques have influenced the development of genetically engineered adenovirus vectors showing therapeutic potential. Increased molecular understanding of the benefits and limitations of HAdVs in preclinical research and clinical studies is a crucial point in the engineering of refined oncolytic vectors. This review presents HAdV species (A-G) used in oncotherapy. We describe the adenovirus genome organizations and modifications, the possibilities oncolytic viruses offer, and their current limitations. Ongoing and ended clinical trials based on oncolytic adenoviruses are presented. This review provides a broad overview of the current knowledge of oncolytic therapy. HAdV-based strategies targeting tumors by employing variable immune modifiers or delivering immune stimulatory factors are of great promise in the field of immune oncologyy This approach can change the face of the fight against cancer, supplying the medical tools to defeat tumors more selectively and safely.
Collapse
Affiliation(s)
- Aleksander Gryciuk
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Marta Rogalska
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Joanna Baran
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH-NRI, 00-791 Warsaw, Poland
- Valo Therapeutics, 00790 Helsinki, Finland
| | - Monika Staniszewska
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| |
Collapse
|
6
|
Najafi S, Majidpoor J, Mortezaee K. The impact of oncolytic adenoviral therapy on the therapeutic efficacy of PD-1/PD-L1 blockade. Biomed Pharmacother 2023; 161:114436. [PMID: 36841031 DOI: 10.1016/j.biopha.2023.114436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/13/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
Immunotherapy has revolutionized treatment of cancer during the last decades. Oncolytic virotherapy has also emerged as a strategy to fight against cancer cells both via lysis of malignant cells and activating immune responses. Accepted as a logical strategy, combination of monoclonal antibodies particularly against the programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) is introduced to improve clinical responses to immune checkpoint inhibitors (ICIs). Accordingly, Talimogene laherparepvec (T-VEC) has received approval for clinical use, while a number of oncolytic Adenoviruses (Ads) are being investigated in clinical trials of malignancies. Combination of oncolytic Ads with PD-1/PD-L1 inhibitors have shown potentials in promoting responses to ICIs, changing the tumor microenvironment, inducing long-term protection against tumor, and promoting survival among mice models of malignancies. Regarding the increasing importance of oncolytic Ads in combination therapy of cancers, in this review we decide to outline recent studies in this field.
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
7
|
Naumenko VA, Stepanenko AA, Lipatova AV, Vishnevskiy DA, Chekhonin VP. Infection of non-cancer cells: A barrier or support for oncolytic virotherapy? MOLECULAR THERAPY - ONCOLYTICS 2022; 24:663-682. [PMID: 35284629 PMCID: PMC8898763 DOI: 10.1016/j.omto.2022.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses are designed to specifically target cancer cells, sparing normal cells. Although numerous studies demonstrate the ability of oncolytic viruses to infect a wide range of non-tumor cells, the significance of this phenomenon for cancer virotherapy is poorly understood. To fill the gap, we summarize the data on infection of non-cancer targets by oncolytic viruses with a special focus on tumor microenvironment and secondary lymphoid tissues. The review aims to address two major questions: how do attenuated viruses manage to infect normal cells, and whether it is of importance for oncolytic virotherapy.
Collapse
Affiliation(s)
- Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Corresponding author Victor A. Naumenko, PhD, V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia.
| | - Aleksei A. Stepanenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
8
|
Sonzogni O, Zak DE, Sasso MS, Lear R, Muntzer A, Zonca M, West K, Champion BR, Rottman JB. T-SIGn tumor reengineering therapy and CAR T cells synergize in combination therapy to clear human lung tumor xenografts and lung metastases in NSG mice. Oncoimmunology 2022; 11:2029070. [PMID: 35154906 PMCID: PMC8837249 DOI: 10.1080/2162402x.2022.2029070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although chimeric antigen receptor (CAR) T cells have emerged as highly effective treatments for patients with hematologic malignancies, similar efficacy has not been achieved in the context of solid tumors. There are several reasons for this disparity including a) fewer solid tumor target antigens, b) heterogenous target expression amongst tumor cells, c) poor trafficking of CAR T cells to the solid tumor and d) an immunosuppressive tumor microenvironment (TME). Oncolytic viruses have the potential to change this paradigm by a) directly lysing tumor cells and releasing tumor neoantigens, b) stimulating the local host innate immune response to release cytokines and recruit additional innate and adaptive immune cells, c) carrying virus-encoded transgenes to “re-program” the TME to a pro-inflammatory environment and d) promoting an adaptive immune response to the neoantigens in this newly permissive TME. Here we show that the Tumor-Specific Immuno-Gene (T-SIGn) virus NG-347 which encodes IFNα, MIP1α and CD80 synergizes with anti-EGFR CAR T cells as well as anti-HER-2 CAR T cells to clear A549 human tumor xenografts and their pulmonary metastases at doses which are subtherapeutic when each is used as a sole treatment. We show that NG-347 changes the TME to a pro-inflammatory environment resulting in the recruitment and activation of both CAR T cells and mouse innate immune cells. We also show that the transgenes encoded by the virus are critical as synergy is lost in their absence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Katy West
- PsiOxus Therapeutics Limited, Abingdon, UK
| | | | | |
Collapse
|
9
|
Liszewski MK, Atkinson JP. Membrane cofactor protein (MCP; CD46): deficiency states and pathogen connections. Curr Opin Immunol 2021; 72:126-134. [PMID: 34004375 PMCID: PMC8123722 DOI: 10.1016/j.coi.2021.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
Membrane cofactor protein (MCP; CD46), a ubiquitously expressed complement regulatory protein, serves as a cofactor for serine protease factor I to cleave and inactivate C3b and C4b deposited on host cells. However, CD46 also plays roles in human reproduction, autophagy, modulating T cell activation and effector functions and is a member of the newly identified intracellular complement system (complosome). CD46 also is a receptor for 11 pathogens ('pathogen magnet'). While CD46 deficiencies contribute to inflammatory disorders, its overexpression in cancers and role as a receptor for some adenoviruses has led to its targeting by oncolytic agents and adenoviral-based therapeutic vectors, including coronavirus disease of 2019 (COVID-19) vaccines. This review focuses on recent advances in identifying disease-causing CD46 variants and its pathogen connections.
Collapse
Affiliation(s)
- M Kathryn Liszewski
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
10
|
Nestić D, Božinović K, Pehar I, Wallace R, Parker AL, Majhen D. The Revolving Door of Adenovirus Cell Entry: Not All Pathways Are Equal. Pharmaceutics 2021; 13:1585. [PMID: 34683878 PMCID: PMC8540258 DOI: 10.3390/pharmaceutics13101585] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
Adenoviruses represent exceptional candidates for wide-ranging therapeutic applications, from vectors for gene therapy to oncolytics for cancer treatments. The first ever commercial gene therapy medicine was based on a recombinant adenovirus vector, while most recently, adenoviral vectors have proven critical as vaccine platforms in effectively controlling the global coronavirus pandemic. Here, we discuss factors involved in adenovirus cell binding, entry, and trafficking; how they influence efficiency of adenovirus-based vectors; and how they can be manipulated to enhance efficacy of genetically modified adenoviral variants. We focus particularly on endocytosis and how different adenovirus serotypes employ different endocytic pathways to gain cell entry, and thus, have different intracellular trafficking pathways that subsequently trigger different host antiviral responses. In the context of gene therapy, the final goal of the adenovirus vector is to efficiently deliver therapeutic transgenes into the target cell nucleus, thus allowing its functional expression. Aberrant or inefficient endocytosis can impede this goal, therefore, it should be considered when designing and constructing adenovirus-based vectors.
Collapse
Affiliation(s)
- Davor Nestić
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| | - Ksenija Božinović
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| | - Isabela Pehar
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| | - Rebecca Wallace
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (R.W.); (A.L.P.)
| | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (R.W.); (A.L.P.)
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (D.N.); (K.B.); (I.P.)
| |
Collapse
|
11
|
Teijeira Crespo A, Burnell S, Capitani L, Bayliss R, Moses E, Mason GH, Davies JA, Godkin AJ, Gallimore AM, Parker AL. Pouring petrol on the flames: Using oncolytic virotherapies to enhance tumour immunogenicity. Immunology 2021; 163:389-398. [PMID: 33638871 PMCID: PMC8274202 DOI: 10.1111/imm.13323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/12/2021] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses possess the ability to infect, replicate and lyse malignantly transformed tumour cells. This oncolytic activity amplifies the therapeutic advantage and induces a form of immunogenic cell death, characterized by increased CD8 + T-cell infiltration into the tumour microenvironment. This important feature of oncolytic viruses can result in the warming up of immunologically 'cold' tumour types, presenting the enticing possibility that oncolytic virus treatment combined with immunotherapies may enhance efficacy. In this review, we assess some of the most promising candidates that might be used for oncolytic virotherapy: immunotherapy combinations. We assess their potential as separate agents or as agents combined into a single therapy, where the immunotherapy is encoded within the genome of the oncolytic virus. The development of such advanced agents will require increasingly sophisticated model systems for their preclinical assessment and evaluation. In vivo rodent model systems are fraught with limitations in this regard. Oncolytic viruses replicate selectively within human cells and therefore require human xenografts in immune-deficient mice for their evaluation. However, the use of immune-deficient rodent models hinders the ability to study immune responses against any immunomodulatory transgenes engineered within the viral genome and expressed within the tumour microenvironment. There has therefore been a shift towards the use of more sophisticated ex vivo patient-derived model systems based on organoids and explant co-cultures with immune cells, which may be more predictive of efficacy than contrived and artificial animal models. We review the best of those model systems here.
Collapse
Affiliation(s)
- Alicia Teijeira Crespo
- Division of Cancer and
GeneticsCardiff University School of Medicine
Cardiff UniversityCardiffUK
| | - Stephanie Burnell
- Division of Infection and Immunity
Cardiff University School of MedicineCardiff UniversityCardiffUK
| | - Lorenzo Capitani
- Division of Infection and Immunity
Cardiff University School of MedicineCardiff UniversityCardiffUK
| | - Rebecca Bayliss
- Division of Cancer and
GeneticsCardiff University School of Medicine
Cardiff UniversityCardiffUK
| | - Elise Moses
- Division of Cancer and
GeneticsCardiff University School of Medicine
Cardiff UniversityCardiffUK
| | - Georgina H. Mason
- Division of Infection and Immunity
Cardiff University School of MedicineCardiff UniversityCardiffUK
| | - James A. Davies
- Division of Cancer and
GeneticsCardiff University School of Medicine
Cardiff UniversityCardiffUK
| | - Andrew J. Godkin
- Division of Infection and Immunity
Cardiff University School of MedicineCardiff UniversityCardiffUK
| | - Awen M. Gallimore
- Division of Infection and Immunity
Cardiff University School of MedicineCardiff UniversityCardiffUK
| | - Alan L. Parker
- Division of Cancer and
GeneticsCardiff University School of Medicine
Cardiff UniversityCardiffUK
| |
Collapse
|
12
|
Atasheva S, Emerson CC, Yao J, Young C, Stewart PL, Shayakhmetov DM. Systemic cancer therapy with engineered adenovirus that evades innate immunity. Sci Transl Med 2021; 12:12/571/eabc6659. [PMID: 33239388 DOI: 10.1126/scitranslmed.abc6659] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
Oncolytic virus therapy is a cancer treatment modality that has the potential to improve outcomes for patients with currently incurable malignancies. Although intravascular delivery of therapeutic viruses provides access to disseminated tumors, this delivery route exposes the virus to opsonizing and inactivating factors in the blood, which limit the effective therapeutic virus dose and contribute to activation of systemic toxicities. When human species C adenovirus HAdv-C5 is delivered intravenously, natural immunoglobulin M (IgM) antibodies and coagulation factor X rapidly opsonize HAdv-C5, leading to virus sequestration in tissue macrophages and promoting infection of liver cells, triggering hepatotoxicity. Here, we showed that natural IgM antibody binds to the hypervariable region 1 (HVR1) of the main HAdv-C5 capsid protein hexon. Using compound targeted mutagenesis of hexon HVR1 loop and other functional sites that mediate virus-host interactions, we engineered and obtained a high-resolution cryo-electron microscopy structure of an adenovirus vector, Ad5-3M, which resisted inactivation by blood factors, avoided sequestration in liver macrophages, and failed to trigger hepatotoxicity after intravenous delivery. Systemic delivery of Ad5-3M to mice with localized or disseminated lung cancer led to viral replication in tumor cells, suppression of tumor growth, and prolonged survival. Thus, compound targeted mutagenesis of functional sites in the virus capsid represents a generalizable approach to tailor virus interactions with the humoral and cellular arms of the immune system, enabling generation of "designer" viruses with improved therapeutic properties.
Collapse
Affiliation(s)
- Svetlana Atasheva
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Corey C Emerson
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jia Yao
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cedrick Young
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Phoebe L Stewart
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA. .,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.,Emory Center for Transplantation and Immune-mediated Disorders, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA.,Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Wan PKT, Ryan AJ, Seymour LW. Beyond cancer cells: Targeting the tumor microenvironment with gene therapy and armed oncolytic virus. Mol Ther 2021; 29:1668-1682. [PMID: 33845199 PMCID: PMC8116634 DOI: 10.1016/j.ymthe.2021.04.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/08/2021] [Accepted: 04/06/2021] [Indexed: 01/17/2023] Open
Abstract
Cancer gene therapies are usually designed either to express wild-type copies of tumor suppressor genes or to exploit tumor-associated phenotypic changes to endow selective cytotoxicity. However, these approaches become less relevant to cancers that contain many independent mutations, and the situation is made more complex by our increased understanding of clonal evolution of tumors, meaning that different metastases and even regions of the same tumor mass have distinct mutational and phenotypic profiles. In contrast, the relatively genetically stable tumor microenvironment (TME) therefore provides an appealing therapeutic target, particularly since it plays an essential role in promoting cancer growth, immune tolerance, and acquired resistance to many therapies. Recently, a variety of different TME-targeted gene therapy and armed oncolytic strategies have been explored, with particular success observed in strategies targeting the cancer stroma, reducing tumor vasculature, and repolarizing the immunosuppressive microenvironment. Herein, we review the progress of these TME-targeting approaches and try to highlight those showing the greatest promise.
Collapse
Affiliation(s)
| | - Anderson J Ryan
- Department Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | | |
Collapse
|
14
|
Heidbuechel JPW, Engeland CE. Oncolytic viruses encoding bispecific T cell engagers: a blueprint for emerging immunovirotherapies. J Hematol Oncol 2021; 14:63. [PMID: 33863363 PMCID: PMC8052795 DOI: 10.1186/s13045-021-01075-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/30/2021] [Indexed: 02/08/2023] Open
Abstract
Bispecific T cell engagers (BiTEs) are an innovative class of immunotherapeutics that redirect T cells to tumor surface antigens. While efficacious against certain hematological malignancies, limited bioavailability and severe toxicities have so far hampered broader clinical application, especially against solid tumors. Another emerging cancer immunotherapy are oncolytic viruses (OVs) which selectively infect and replicate in malignant cells, thereby mediating tumor vaccination effects. These oncotropic viruses can serve as vectors for tumor-targeted immunomodulation and synergize with other immunotherapies. In this article, we discuss the use of OVs to overcome challenges in BiTE therapy. We review the current state of the field, covering published preclinical studies as well as ongoing clinical investigations. We systematically introduce OV-BiTE vector design and characteristics as well as evidence for immune-stimulating and anti-tumor effects. Moreover, we address additional combination regimens, including CAR T cells and immune checkpoint inhibitors, and further strategies to modulate the tumor microenvironment using OV-BiTEs. The inherent complexity of these novel therapeutics highlights the importance of translational research including correlative studies in early-phase clinical trials. More broadly, OV-BiTEs can serve as a blueprint for diverse OV-based cancer immunotherapies.
Collapse
Affiliation(s)
- Johannes P W Heidbuechel
- Research Group Mechanisms of Oncolytic Immunotherapy, Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Christine E Engeland
- Research Group Mechanisms of Oncolytic Immunotherapy, Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
- Department of Medical Oncology, University Hospital Heidelberg, Heidelberg, Germany.
- Center for Biomedical Research and Education (ZBAF), School of Medicine, Institute of Virology and Microbiology, Faculty of Health, Witten/Herdecke University, Witten, Germany.
| |
Collapse
|
15
|
Piwoni K, Jaeckel G, Rasa A, Alberts P. 4-Week repeated dose rat GLP toxicity study of oncolytic ECHO-7 virus Rigvir administered intramuscularly with a 4-week recovery period. Toxicol Rep 2021; 8:230-238. [PMID: 33537211 PMCID: PMC7840795 DOI: 10.1016/j.toxrep.2021.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 11/24/2022] Open
Abstract
4-week repeated dose rat GLP toxicology study with the oncolytic ECHO-7 virus Rigvir. No unscheduled deaths, adverse clinical signs, changes in body weight or food intake. Rigvir was biodistributed to the spleen. Rigvir was well tolerated in the rats. The no-observed-adverse-effect level (NOAEL) was the highest dose tested 2×107 TCID50.
The oncolytic ECHO-7 virus Rigvir was registered in Latvia in 2004 and later in Georgia, Armenia and Uzbekistan. No severe adverse events have been observed. During drug development good laboratory practice (GLP) pre-clinical toxicology studies are generally required by regulatory agencies. Since such studies had previously not been performed, the aim of this 4-week repeated dose GLP toxicity study was to determine the potential toxicity, and reversibility of any findings after a 4-week treatment-free period. Han-Wistar rats were randomly assigned to control, Rigvir (2×106, 1×107 and 2×107 TCID50) groups. Intramuscular administration was on days 1-3, 8-10, 15-17, and 22-24. Clinical signs, average food-intake, body weights, ophthalmology, clinical pathology parameters, bioanalysis, gross necropsy, organ weights, biodistribution and histopathology were evaluated. There were no unscheduled deaths, adverse clinical signs, no changes in body weight, body weight gain, food intake, ophthalmoscopy, clinical pathology, urine volume or composition, or organ weights. Slightly higher numbers of eosinophils in Rigvir treated animals returned to normal after recovery. Rigvir biodistributed to the spleen. Low incidence of inflammatory cell infiltration at administration sites and increased lymphoid cellularity at the regional (inguinal and popliteal) lymph nodes were observed; after recovery, only those in popliteal lymph nodes remained. Therefore, 4-week Rigvir at 2×107 TCID50 administration was well tolerated in rats. The no-observed-adverse-effect level (NOAEL) was the highest dose tested, 2×107 TCID50. Objectives The objectives of this study were to determine the potential toxicity of Rigvir, an ECHO-7 oncolytic virus, when administered intramuscularly for 4 weeks to rats, with a 4-week recovery period, and to evaluate the reversibility of any potential findings. In addition, the biodistribution of Rigvir in selected tissues was determined.
Collapse
Affiliation(s)
- Katarzyna Piwoni
- Charles River Laboratories Edinburgh Ltd., Elphinstone Research Centre Tranent, East Lothian, EH33 2NE, UK
| | - Gilta Jaeckel
- Charles River Laboratories Edinburgh Ltd., Elphinstone Research Centre Tranent, East Lothian, EH33 2NE, UK
| | - Agnija Rasa
- Rigvir, Riga, Latvia
- Corresponding author at: Rigvir, Atlasa iela 7C, Riga, LV-1026, Latvia.
| | | |
Collapse
|
16
|
Hwang JK, Hong J, Yun CO. Oncolytic Viruses and Immune Checkpoint Inhibitors: Preclinical Developments to Clinical Trials. Int J Mol Sci 2020; 21:E8627. [PMID: 33207653 PMCID: PMC7697902 DOI: 10.3390/ijms21228627] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Immuno-oncology (IO) has been an active area of oncology research. Following US FDA approval of the first immune checkpoint inhibitor (ICI), ipilimumab (human IgG1 k anti-CTLA-4 monoclonal antibody), in 2011, and of the first oncolytic virus, Imlygic (talimogene laherparepvec), in 2015, there has been renewed interest in IO. In the past decade, ICIs have changed the treatment paradigm for many cancers by enabling better therapeutic control, resuming immune surveillance, suppressing tumor immunosuppression, and restoring antitumor immune function. However, ICI therapies are effective only in a small subset of patients and show limited therapeutic potential due to their inability to demonstrate efficacy in 'cold' or unresponsive tumor microenvironments (TMEs). Relatedly, oncolytic viruses (OVs) have been shown to induce antitumor immune responses, augment the efficacy of existing cancer treatments, and reform unresponsive TME to turn 'cold' tumors 'hot,' increasing their susceptibility to checkpoint blockade immunotherapies. For this reason, OVs serve as ideal complements to ICIs, and multiple preclinical studies and clinical trials are demonstrating their combined therapeutic efficacy. This review will discuss the merits and limitations of OVs and ICIs as monotherapy then progress onto the preclinical rationale and the results of clinical trials of key combination therapies.
Collapse
Affiliation(s)
- June Kyu Hwang
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea; (J.K.H.); (J.H.)
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea; (J.K.H.); (J.H.)
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea; (J.K.H.); (J.H.)
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
- Institute of Nano Science and Technology, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
| |
Collapse
|
17
|
CD46 and Oncologic Interactions: Friendly Fire against Cancer. Antibodies (Basel) 2020; 9:antib9040059. [PMID: 33147799 PMCID: PMC7709105 DOI: 10.3390/antib9040059] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/14/2020] [Accepted: 10/25/2020] [Indexed: 12/16/2022] Open
Abstract
One of the most challenging aspects of cancer therapeutics is target selection. Recently, CD46 (membrane cofactor protein; MCP) has emerged as a key player in both malignant transformation as well as in cancer treatments. Normally a regulator of complement activation, CD46 is co-expressed as four predominant isoforms on almost all cell types. CD46 is highly overexpressed on a variety of human tumor cells. Clinical and experimental data support an association between increased CD46 expression and malignant transformation and metastasizing potential. Further, CD46 is a newly discovered driver of metabolic processes and plays a role in the intracellular complement system (complosome). CD46 is also known as a pathogen magnet due to its role as a receptor for numerous microbes, including several species of measles virus and adenoviruses. Strains of these two viruses have been exploited as vectors for the therapeutic development of oncolytic agents targeting CD46. In addition, monoclonal antibody-drug conjugates against CD46 also are being clinically evaluated. As a result, there are multiple early-phase clinical trials targeting CD46 to treat a variety of cancers. Here, we review CD46 relative to these oncologic connections.
Collapse
|
18
|
Challenges and strategies for next-generation bispecific antibody-based antitumor therapeutics. Cell Mol Immunol 2020; 17:451-461. [PMID: 32313210 DOI: 10.1038/s41423-020-0417-8] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Bispecific antibodies (bsAbs) refer to a large family of molecules that recognize two different epitopes or antigens. Although a series of challenges, especially immunogenicity and chain mispairing issues, once hindered the development of bsAbs, they have been gradually overcome with the help of rapidly developing technologies in the past 5 decades. In the meantime, an increasing number of bsAb platforms have been designed to satisfy different clinical demands. Currently, numerous preclinical and clinical trials are underway, portraying a promising future for bsAb-based cancer treatment. Nevertheless, bsAb drugs still face enormous challenges in their application as cancer therapeutics, including tumor heterogeneity and mutational burden, intractable tumor microenvironment (TME), insufficient costimulatory signals to activate T cells, the necessity for continuous injection, fatal systemic side effects, and off-target toxicities to adjacent normal cells. Therefore, we provide several strategies as solutions to these issues, which comprise generating multispecific bsAbs, discovering neoantigens, combining bsAbs with other anticancer therapies, exploiting natural killer (NK)-cell-based bsAbs and producing bsAbs in situ. In this review, we mainly discuss previous and current challenges in bsAb development and underscore corresponding strategies, with a brief introduction of several typical bsAb formats.
Collapse
|
19
|
Yousaf I, Kaeppler J, Frost S, Seymour LW, Jacobus EJ. Attenuation of the Hypoxia Inducible Factor Pathway after Oncolytic Adenovirus Infection Coincides with Decreased Vessel Perfusion. Cancers (Basel) 2020; 12:E851. [PMID: 32244697 PMCID: PMC7225929 DOI: 10.3390/cancers12040851] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/30/2022] Open
Abstract
The interplay between oncolytic virus infection and tumour hypoxia is particularly unexplored in vivo, although hypoxia is present in virtually all solid carcinomas. In this study, oncolytic adenovirus infection foci were found within pimonidazole-reactive, oxygen-poor areas in a colorectal xenograft tumour, where the expression of VEGF, a target gene of the hypoxia-inducible factor (HIF), was attenuated. We hypothesised that adenovirus infection interferes with the HIF-signalling axis in the hypoxic tumour niche, possibly modifying the local vascular supply. In vitro, enadenotucirev (EnAd), adenovirus 11p and adenovirus 5 decreased the protein expression of HIF-1α only during the late phase of the viral life cycle by transcriptional down-regulation and not post-translational regulation. The decreasing HIF levels resulted in the down-regulation of angiogenic factors such as VEGF, coinciding with reduced endothelial tube formation but also increased T-cell activation in conditioned media transfer experiments. Using intravital microscopy, a decreased perfused vessel volume was observed in infected tumour nodules upon systemic delivery of EnAd, encoding the oxygen-independent fluorescent reporter UnaG to a tumour xenograft grown under an abdominal window chamber. We conclude that the attenuation of the HIF pathway upon adenoviral infection may contribute to anti-vascular and immunostimulatory effects in the periphery of established infection foci in vivo.
Collapse
Affiliation(s)
- Iris Yousaf
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (I.Y.); (S.F.)
| | - Jakob Kaeppler
- Mechanisms of Metastasis Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK;
| | - Sally Frost
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (I.Y.); (S.F.)
| | - Len W. Seymour
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (I.Y.); (S.F.)
| | - Egon J. Jacobus
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (I.Y.); (S.F.)
| |
Collapse
|
20
|
Leung EY, Ennis DP, Kennedy PR, Hansell C, Dowson S, Farquharson M, Spiliopoulou P, Nautiyal J, McNamara S, Carlin LM, Fisher K, Davis DM, Graham G, McNeish IA. NK Cells Augment Oncolytic Adenovirus Cytotoxicity in Ovarian Cancer. Mol Ther Oncolytics 2020; 16:289-301. [PMID: 32195317 PMCID: PMC7068056 DOI: 10.1016/j.omto.2020.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/10/2020] [Indexed: 12/20/2022] Open
Abstract
Oncolytic viruses (OVs) can trigger profound innate and adaptive immune responses, which have the potential both to potentiate and reduce the activity of OVs. Natural killer (NK) cells can mediate potent anti-viral and anti-tumoral responses, but there are no data on the role of NK cells in oncolytic adenovirus activity. Here, we have used two different oncolytic adenoviruses-the Ad5 E1A CR2-deletion mutant dl922-947 (group C) and the chimeric Ad3/Ad11p mutant enadenotucirev (group B)-to investigate the effect of NK cells on overall anti-cancer efficacy in ovarian cancer. Because human adenoviruses do not replicate in murine cells, we utilized primary human NK cells from peripheral blood and ovarian cancer ascites. Our results show that dl922-947 and enadenotucirev do not infect NK cells, but induce contact-dependent activation and anti-cancer cytotoxicity against adenovirus-infected ovarian cancer cells. Moreover, manipulation of NK receptors DNAM-1 (DNAX accessory molecule-1) and TIGIT (T cell immunoreceptor with Ig and ITIM domains) significantly influences NK cytotoxicity against adenovirus-infected cells. Together, these results indicate that NK cells act to increase the activity of oncolytic adenovirus in ovarian cancer and suggest that strategies to augment NK activity further via the blockade of inhibitory NK receptor TIGIT could enhance therapeutic potential of OVs.
Collapse
Affiliation(s)
- Elaine Y.L. Leung
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Institute of Infection, Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Darren P. Ennis
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Ovarian Cancer Action Research Centre and Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
| | - Philippa R. Kennedy
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Christopher Hansell
- Institute of Infection, Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Suzanne Dowson
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Pavlina Spiliopoulou
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Ovarian Cancer Action Research Centre and Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
| | - Jaya Nautiyal
- Ovarian Cancer Action Research Centre and Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
| | - Sophie McNamara
- Ovarian Cancer Action Research Centre and Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
| | | | | | - Daniel M. Davis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Gerard Graham
- Institute of Infection, Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Iain A. McNeish
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Ovarian Cancer Action Research Centre and Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
| |
Collapse
|
21
|
Pokrovska TD, Jacobus EJ, Puliyadi R, Prevo R, Frost S, Dyer A, Baugh R, Rodriguez-Berriguete G, Fisher K, Granata G, Herbert K, Taverner WK, Champion BR, Higgins GS, Seymour LW, Lei-Rossmann J. External Beam Radiation Therapy and Enadenotucirev: Inhibition of the DDR and Mechanisms of Radiation-Mediated Virus Increase. Cancers (Basel) 2020; 12:E798. [PMID: 32224979 PMCID: PMC7226394 DOI: 10.3390/cancers12040798] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 11/17/2022] Open
Abstract
Ionising radiation causes cell death through the induction of DNA damage, particularly double-stranded DNA (dsDNA) breaks. Evidence suggests that adenoviruses inhibit proteins involved in the DNA damage response (DDR) to prevent recognition of double-stranded viral DNA genomes as cellular dsDNA breaks. We hypothesise that combining adenovirus treatment with radiotherapy has the potential for enhancing tumour-specific cytotoxicity through inhibition of the DDR and augmentation of virus production. We show that EnAd, an Ad3/Ad11p chimeric oncolytic adenovirus currently being trialled in colorectal and other cancers, targets the DDR pathway at a number of junctures. Infection is associated with a decrease in irradiation-induced 53BP1 and Rad51 foci formation, and in total DNA ligase IV levels. We also demonstrate a radiation-associated increase in EnAd production in vitro and in a pilot in vivo experiment. Given the current limitations of in vitro techniques in assessing for synergy between these treatments, we adapted the plaque assay to allow monitoring of viral plaque size and growth and utilised the xCELLigence cell adhesion assay to measure cytotoxicity. Our study provides further evidence on the interaction between adenovirus and radiation in vitro and in vivo and suggests these have at least an additive, and possibly a synergistic, impact on cytotoxicity.
Collapse
Affiliation(s)
- Tzveta D. Pokrovska
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | - Egon J. Jacobus
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | - Rathi Puliyadi
- Tumour Radiosensitivity Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.P.); (R.P.); (G.R.-B.); (G.G.); (K.H.); (G.S.H.)
| | - Remko Prevo
- Tumour Radiosensitivity Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.P.); (R.P.); (G.R.-B.); (G.G.); (K.H.); (G.S.H.)
| | - Sally Frost
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | - Arthur Dyer
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | - Richard Baugh
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | - Gonzalo Rodriguez-Berriguete
- Tumour Radiosensitivity Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.P.); (R.P.); (G.R.-B.); (G.G.); (K.H.); (G.S.H.)
| | - Kerry Fisher
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
- PsiOxus Therapeutics Ltd., Abingdon OX14 3YS, UK;
| | - Giovanna Granata
- Tumour Radiosensitivity Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.P.); (R.P.); (G.R.-B.); (G.G.); (K.H.); (G.S.H.)
| | - Katharine Herbert
- Tumour Radiosensitivity Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.P.); (R.P.); (G.R.-B.); (G.G.); (K.H.); (G.S.H.)
| | - William K. Taverner
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | | | - Geoff S. Higgins
- Tumour Radiosensitivity Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.P.); (R.P.); (G.R.-B.); (G.G.); (K.H.); (G.S.H.)
| | - Len W. Seymour
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| | - Janet Lei-Rossmann
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (T.D.P.); (E.J.J.); (S.F.); (A.D.); (R.B.); (K.F.); (W.K.T.); (J.L.-R.)
| |
Collapse
|
22
|
Taverner WK, Jacobus EJ, Christianson J, Champion B, Paton AW, Paton JC, Su W, Cawood R, Seymour LW, Lei-Rossmann J. Calcium Influx Caused by ER Stress Inducers Enhances Oncolytic Adenovirus Enadenotucirev Replication and Killing through PKCα Activation. Mol Ther Oncolytics 2019; 15:117-130. [PMID: 31890865 PMCID: PMC6931121 DOI: 10.1016/j.omto.2019.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/22/2019] [Indexed: 01/17/2023] Open
Abstract
Oncolytic viruses represent an emerging approach to cancer therapy. However, better understanding of their interaction with the host cancer cell and approaches to enhance their efficacy are needed. Here, we investigate the effect of chemically induced endoplasmic reticulum (ER) stress on the activity of the chimeric group B adenovirus Enadenotucirev, its closely related parental virus Ad11p, and the archetypal group C oncolytic adenovirus Ad5. We show that treatment of colorectal and ovarian cancer cell lines with thapsigargin or ionomycin caused an influx of Ca2+, leading to an upregulation in E1A transcript and protein levels. Increased E1A protein levels, in turn, increased levels of expression of the E2B viral DNA polymerase, genome replication, late viral protein expression, infectious virus particle production, and cell killing during Enadenotucirev and Ad11p, but not Ad5, infection. This effect was not due to the induction of ER stress, but rather the influx of extracellular Ca2+ and consequent increase in protein kinase C activity. These results underscore the importance of Ca2+ homeostasis during adenoviral infection, indicate a signaling pathway between protein kinase C and E1A, and raise the possibility of using Ca2+ flux-modulating agents in the manufacture and potentiation of oncolytic virotherapies.
Collapse
Affiliation(s)
- William K. Taverner
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Egon J. Jacobus
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - John Christianson
- NDORMS, Botnar Research Centre, University of Oxford, Headington, Oxford OX3 7LD, UK
| | - Brian Champion
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 3YS, UK
| | - Adrienne W. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide SA 5005, Australia
| | - James C. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide SA 5005, Australia
| | - Weiheng Su
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Ryan Cawood
- Oxford Genetics Ltd., Medawar Centre, Robert Robinson Avenue, Oxford OX4 4HG, UK
| | - Len W. Seymour
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Janet Lei-Rossmann
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
23
|
Atasheva S, Yao J, Shayakhmetov DM. Innate immunity to adenovirus: lessons from mice. FEBS Lett 2019; 593:3461-3483. [PMID: 31769012 PMCID: PMC6928416 DOI: 10.1002/1873-3468.13696] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/07/2019] [Accepted: 11/21/2019] [Indexed: 01/01/2023]
Abstract
Adenovirus is a highly evolutionary successful pathogen, as it is widely prevalent across the animal kingdom, infecting hosts ranging from lizards and frogs to dolphins, birds, and humans. Although natural adenovirus infections in humans rarely cause severe pathology, intravenous injection of high doses of adenovirus-based vectors triggers rapid activation of the innate immune system, leading to cytokine storm syndrome, disseminated intravascular coagulation, thrombocytopenia, and hepatotoxicity, which individually or in combination may cause morbidity and mortality. Much of the information on exactly how adenovirus activates the innate immune system has been gathered from mouse experimental systems. Intravenous administration of adenovirus to mice revealed mechanistic insights into cellular and molecular components of the innate immunity that detect adenovirus particles, activate pro-inflammatory signaling pathways and cytokine production, sequester adenovirus particles from the bloodstream, and eliminate adenovirus-infected cells. Collectively, this information greatly improved our understanding of mechanisms of activation of innate immunity to adenovirus and may pave the way for designing safer adenovirus-based vectors for therapy of genetic and acquired human diseases.
Collapse
Affiliation(s)
- Svetlana Atasheva
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jia Yao
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dmitry M. Shayakhmetov
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Children’s Center for Transplantation and Immuno-mediated Disorders, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
24
|
Scott EM, Jacobus EJ, Lyons B, Frost S, Freedman JD, Dyer A, Khalique H, Taverner WK, Carr A, Champion BR, Fisher KD, Seymour LW, Duffy MR. Bi- and tri-valent T cell engagers deplete tumour-associated macrophages in cancer patient samples. J Immunother Cancer 2019; 7:320. [PMID: 31753017 PMCID: PMC6873687 DOI: 10.1186/s40425-019-0807-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Tumour-associated macrophages (TAMs) are often implicated in cancer progression but can also exert anti-tumour activities. Selective eradication of cancer-promoting (M2-like) TAM subsets is a highly sought-after goal. Here, we have devised a novel strategy to achieve selective TAM depletion, involving the use of T cell engagers to direct endogenous T cell cytotoxicity towards specific M2-like TAMs. To avoid "on-target off-tumour" toxicities, we have explored localising expression of the T cell engagers to the tumour with enadenotucirev (EnAd), an oncolytic adenovirus in Phase I/II clinical trials. METHOD A panel of bi- and tri-valent T cell engagers (BiTEs/TriTEs) was constructed, recognising CD3ε on T cells and CD206 or folate receptor β (FRβ) on M2-like macrophages. Initial characterisation of BiTE/TriTE activity and specificity was performed with M1- and M2-polarised monocyte-derived macrophages and autologous lymphocytes from healthy human peripheral blood donors. T cell engagers were inserted into the genome of EnAd, and oncolytic activity and BiTE secretion assessed with DLD-1 tumour cells. Clinically-relevant ex vivo models (whole malignant ascites from cancer patients) were employed to assess the efficacies of the free- and virally-encoded T cell engagers. RESULTS T cells activated by the CD206- and FRβ-targeting BiTEs/TriTEs preferentially killed M2- over M1-polarised autologous macrophages, with EC50 values in the nanomolar range. A TriTE with bivalent CD3ε binding - the first of its kind - demonstrated enhanced potency whilst retaining target cell selectivity, whereas a CD28-containing TriTE elicited non-specific T cell activation. In immunosuppressive malignant ascites, both free and EnAd-encoded T cell engagers triggered endogenous T cell activation and IFN-γ production, leading to increased T cell numbers and depletion of CD11b+CD64+ ascites macrophages. Strikingly, surviving macrophages exhibited a general increase in M1 marker expression, suggesting microenvironmental repolarisation towards a pro-inflammatory state. CONCLUSIONS This study is the first to achieve selective depletion of specific M2-like macrophage subsets, opening the possibility of eradicating cancer-supporting TAMs whilst sparing those with anti-tumour potential. Targeted TAM depletion with T cell engager-armed EnAd offers a powerful therapeutic approach combining direct cancer cell cytotoxicity with reversal of immune suppression.
Collapse
Affiliation(s)
- Eleanor M. Scott
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Egon J. Jacobus
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Brian Lyons
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Sally Frost
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | | | - Arthur Dyer
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Hena Khalique
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | | | - Alison Carr
- Churchill Hospital, Oxford University Hospital NHS Trust, Oxford, OX3 7LE UK
| | | | - Kerry D. Fisher
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | - Len W. Seymour
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ UK
| | | |
Collapse
|
25
|
CryoEM structure of adenovirus type 3 fibre with desmoglein 2 shows an unusual mode of receptor engagement. Nat Commun 2019; 10:1181. [PMID: 30862836 PMCID: PMC6414520 DOI: 10.1038/s41467-019-09220-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/26/2019] [Indexed: 11/09/2022] Open
Abstract
Attachment of human adenovirus (HAd) to the host cell is a critical step of infection. Initial attachment occurs via the adenoviral fibre knob protein and a cellular receptor. Here we report the cryo-electron microscopy (cryo-EM) structure of a <100 kDa non-symmetrical complex comprising the trimeric HAd type 3 fibre knob (HAd3K) and human desmoglein 2 (DSG2). The structure reveals a unique stoichiometry of 1:1 and 2:1 (DSG2: knob trimer) not previously observed for other HAd-receptor complexes. We demonstrate that mutating Asp261 in the fibre knob is sufficient to totally abolish receptor binding. These data shed new light on adenovirus infection strategies and provide insights for adenoviral vector development and structure-based design.
Collapse
|
26
|
Baker AT, Greenshields-Watson A, Coughlan L, Davies JA, Uusi-Kerttula H, Cole DK, Rizkallah PJ, Parker AL. Diversity within the adenovirus fiber knob hypervariable loops influences primary receptor interactions. Nat Commun 2019; 10:741. [PMID: 30765704 PMCID: PMC6376029 DOI: 10.1038/s41467-019-08599-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/21/2019] [Indexed: 01/14/2023] Open
Abstract
Adenovirus based vectors are of increasing importance for wide ranging therapeutic applications. As vaccines, vectors derived from human adenovirus species D serotypes 26 and 48 (HAdV-D26/48) are demonstrating promising efficacy as protective platforms against infectious diseases. Significant clinical progress has been made, yet definitive studies underpinning mechanisms of entry, infection, and receptor usage are currently lacking. Here, we perform structural and biological analysis of the receptor binding fiber-knob protein of HAdV-D26/48, reporting crystal structures, and modelling putative interactions with two previously suggested attachment receptors, CD46 and Coxsackie and Adenovirus Receptor (CAR). We provide evidence of a low affinity interaction with CAR, with modelling suggesting affinity is attenuated through extended, semi-flexible loop structures, providing steric hindrance. Conversely, in silico and in vitro experiments are unable to provide evidence of interaction between HAdV-D26/48 fiber-knob with CD46, or with Desmoglein 2. Our findings provide insight into the cell-virus interactions of HAdV-D26/48, with important implications for the design and engineering of optimised Ad-based therapeutics.
Collapse
Affiliation(s)
- Alexander T Baker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | | | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - James A Davies
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Hanni Uusi-Kerttula
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - David K Cole
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.,Immunocore Ltd., Abingdon, OX14 4RY, Oxon, UK
| | - Pierre J Rizkallah
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Alan L Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
| |
Collapse
|
27
|
Wang H, Georgakopoulou A, Psatha N, Li C, Capsali C, Samal HB, Anagnostopoulos A, Ehrhardt A, Izsvák Z, Papayannopoulou T, Yannaki E, Lieber A. In vivo hematopoietic stem cell gene therapy ameliorates murine thalassemia intermedia. J Clin Invest 2019; 129:598-615. [PMID: 30422819 PMCID: PMC6355219 DOI: 10.1172/jci122836] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/06/2018] [Indexed: 12/16/2022] Open
Abstract
Current thalassemia gene therapy protocols require the collection of hematopoietic stem/progenitor cells (HSPCs), in vitro culture, lentivirus vector transduction, and retransplantation into myeloablated patients. Because of cost and technical complexity, it is unlikely that such protocols will be applicable in developing countries, where the greatest demand for a β-thalassemia therapy lies. We have developed a simple in vivo HSPC gene therapy approach that involves HSPC mobilization and an intravenous injection of integrating HDAd5/35++ vectors. Transduced HSPCs homed back to the bone marrow, where they persisted long-term. HDAd5/35++ vectors for in vivo gene therapy of thalassemia had a unique capsid that targeted primitive HSPCs through human CD46, a relatively safe SB100X transposase-based integration machinery, a micro-LCR-driven γ-globin gene, and an MGMT(P140K) system that allowed for increasing the therapeutic effect by short-term treatment with low-dose O6-benzylguanine plus bis-chloroethylnitrosourea. We showed in "healthy" human CD46-transgenic mice and in a mouse model of thalassemia intermedia that our in vivo approach resulted in stable γ-globin expression in the majority of circulating red blood cells. The high marking frequency was maintained in secondary recipients. In the thalassemia model, a near-complete phenotypic correction was achieved. The treatment was well tolerated. This cost-efficient and "portable" approach could permit a broader clinical application of thalassemia gene therapy.
Collapse
Affiliation(s)
- Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Aphrodite Georgakopoulou
- Gene and Cell Therapy Center, Hematology Department, George Papanicolaou Hospital, Thessaloniki, Greece
- Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikoletta Psatha
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Chrysi Capsali
- Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Achilles Anagnostopoulos
- Gene and Cell Therapy Center, Hematology Department, George Papanicolaou Hospital, Thessaloniki, Greece
| | | | | | | | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department, George Papanicolaou Hospital, Thessaloniki, Greece
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
28
|
Machiels JP, Salazar R, Rottey S, Duran I, Dirix L, Geboes K, Wilkinson-Blanc C, Pover G, Alvis S, Champion B, Fisher K, McElwaine-Johnn H, Beadle J, Calvo E. A phase 1 dose escalation study of the oncolytic adenovirus enadenotucirev, administered intravenously to patients with epithelial solid tumors (EVOLVE). J Immunother Cancer 2019; 7:20. [PMID: 30691536 PMCID: PMC6348630 DOI: 10.1186/s40425-019-0510-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/13/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Enadenotucirev is a chimeric adenovirus with demonstrated preclinical tumor-selective cytotoxicity and a short half-life. Further clinical mechanism of action data showed that enadenotucirev can gain access to and replicate within different types of epithelial tumors. This phase 1 dose escalation study assessed intravenous (IV) dose escalation with enadenotucirev to establish the maximum tolerated dose (MTD) and subsequently identify a suitable schedule for repeated cycles. METHODS Sixty-one patients with advanced epithelial tumors unresponsive to conventional therapy were enrolled and received enadenotucirev monotherapy as part of this study. During the phase 1a dose escalation (n = 22) and expansion (n = 9), delivery of enadenotucirev between 1 × 1010 and 1 × 1013 viral particles (vp) on days 1, 3, and 5 (single cycle) was used to determine an appropriate MTD. Subsequent treatment cohorts (phase 1a, n = 6 and phase 1b, n = 24) examined the feasibility of repeated dosing cycles in either 3-weekly or weekly dosing regimens. RESULTS Enadenotucirev displayed a predictable and manageable safety profile at doses up to the MTD of 3 × 1012 vp, irrespective of infusion time or dosing schedule. The most commonly reported treatment-emergent adverse events (TEAEs) of grade 3 or higher were hypoxia, lymphopenia, and neutropenia. The frequency of all TEAEs (notably pyrexia and chills) was highest within 24 h of the first enadenotucirev infusion and decreased upon subsequent dosing. Additionally, delivery of three doses of enadenotucirev over 5 days optimized pharmacokinetic and chemokine profiles in the circulation over time. CONCLUSIONS This study provides key clinical data in patients with solid epithelial tumors following treatment with IV enadenotucirev monotherapy and supports further investigation of enadenotucirev in combination with other therapeutic agents at doses up to the MTD of 3 × 1012 vp. TRIAL REGISTRATION ( ClinicalTrials.gov Identifier: NCT02028442 ). Trial registration date: 07 January 2014 - Retrospectively registered.
Collapse
Affiliation(s)
- Jean-Pascal Machiels
- Department of Medical Oncology, Institut Roi Albert II, Cliniques universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale, Université catholique de Louvain, Brussels, Belgium
| | - Ramon Salazar
- Medical Oncology Department, Catalan Institute of Oncology, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Sylvie Rottey
- Drug Research Unit Ghent, Ghent University Hospital, Ghent, Belgium
| | - Ignacio Duran
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Luc Dirix
- Saint-Augustinus Hospital, Antwerp, Belgium
| | - Karen Geboes
- Department of Gastroenterology and Digestive Oncology, Ghent University Hospital, Ghent, Belgium
| | | | - Gillian Pover
- PsiOxus Therapeutics Limited, 4-10 The Quadrant, Barton Lane, Abingdon, UK
| | - Simon Alvis
- PsiOxus Therapeutics Limited, 4-10 The Quadrant, Barton Lane, Abingdon, UK
| | - Brian Champion
- PsiOxus Therapeutics Limited, 4-10 The Quadrant, Barton Lane, Abingdon, UK.
| | - Kerry Fisher
- PsiOxus Therapeutics Limited, 4-10 The Quadrant, Barton Lane, Abingdon, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - John Beadle
- PsiOxus Therapeutics Limited, 4-10 The Quadrant, Barton Lane, Abingdon, UK
| | - Emiliano Calvo
- START Madrid, Centro Integral Oncológico Clara Campal, Hospital Madrid Norte Sanchinarro, Madrid, Spain
| |
Collapse
|
29
|
Freedman JD, Duffy MR, Lei-Rossmann J, Muntzer A, Scott EM, Hagel J, Campo L, Bryant RJ, Verrill C, Lambert A, Miller P, Champion BR, Seymour LW, Fisher KD. An Oncolytic Virus Expressing a T-cell Engager Simultaneously Targets Cancer and Immunosuppressive Stromal Cells. Cancer Res 2018; 78:6852-6865. [PMID: 30449733 DOI: 10.1158/0008-5472.can-18-1750] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/18/2018] [Accepted: 10/16/2018] [Indexed: 11/16/2022]
Abstract
: Effective immunotherapy of stromal-rich tumors requires simultaneous targeting of cancer cells and immunosuppressive elements of the microenvironment. Here, we modified the oncolytic group B adenovirus enadenotucirev to express a stroma-targeted bispecific T-cell engager (BiTE). This BiTE bound fibroblast activation protein on cancer-associated fibroblasts (CAF) and CD3ε on T cells, leading to potent T-cell activation and fibroblast death. Treatment of fresh clinical biopsies, including malignant ascites and solid prostate cancer tissue, with FAP-BiTE-encoding virus induced activation of tumor-infiltrating PD1+ T cells to kill CAFs. In ascites, this led to depletion of CAF-associated immunosuppressive factors, upregulation of proinflammatory cytokines, and increased gene expression of markers of antigen presentation, T-cell function, and trafficking. M2-like ascites macrophages exhibited a proinflammatory repolarization, indicating spectrum-wide alteration of the tumor microenvironment. With this approach, we have actively killed both cancer cells and tumor fibroblasts, reversing CAF-mediated immunosuppression and yielding a potent single-agent therapeutic that is ready for clinical assessment. SIGNIFICANCE: An engineered oncolytic adenovirus that encodes a bispecific antibody combines direct virolysis with endogenous T-cell activation to attack stromal fibroblasts, providing a multimodal treatment strategy within a single therapeutic agent.
Collapse
Affiliation(s)
- Joshua D Freedman
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Margaret R Duffy
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | | | - Eleanor M Scott
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Joachim Hagel
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Leticia Campo
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Richard J Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Clare Verrill
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Adam Lambert
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Paul Miller
- Churchill Hospital, Oxford University Hospital NHS Trust, Oxford, United Kingdom
| | | | - Leonard W Seymour
- Department of Oncology, University of Oxford, Oxford, United Kingdom.
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Emerging functional markers for cancer stem cell-based therapies: Understanding signaling networks for targeting metastasis. Semin Cancer Biol 2018; 53:90-109. [PMID: 29966677 DOI: 10.1016/j.semcancer.2018.06.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Metastasis is one of the most challenging issues in cancer patient management, and effective therapies to specifically target disease progression are missing, emphasizing the urgent need for developing novel anti-metastatic therapeutics. Cancer stem cells (CSCs) gained fast attention as a minor population of highly malignant cells within liquid and solid tumors that are responsible for tumor onset, self-renewal, resistance to radio- and chemotherapies, and evasion of immune surveillance accelerating recurrence and metastasis. Recent progress in the identification of their phenotypic and molecular characteristics and interactions with the tumor microenvironment provides great potential for the development of CSC-based targeted therapies and radical improvement in metastasis prevention and cancer patient prognosis. Here, we report on newly uncovered signaling mechanisms controlling CSC's aggressiveness and treatment resistance, and CSC-specific agents and molecular therapeutics, some of which are currently under investigation in clinical trials, gearing towards decisive functional CSC intrinsic or surface markers. One special research focus rests upon subverted regulatory pathways such as insulin-like growth factor 1 receptor signaling and its interactors in metastasis-initiating cell populations directly related to the gain of stem cell- and EMT-associated properties, as well as key components of the E2F transcription factor network regulating metastatic progression, microenvironmental changes, and chemoresistance. In addition, the study provides insight into systems biology tools to establish complex molecular relationships behind the emergence of aggressive phenotypes from high-throughput data that rely on network-based analysis and their use to investigate immune escape mechanisms or predict clinical outcome-relevant CSC receptor signaling signatures. We further propose that customized vector technologies could drastically enhance systemic drug delivery to target sites, and summarize recent progress and remaining challenges. This review integrates available knowledge on CSC biology, computational modeling approaches, molecular targeting strategies, and delivery techniques to envision future clinical therapies designed to conquer metastasis-initiating cells.
Collapse
|
31
|
Baker AT, Aguirre-Hernández C, Halldén G, Parker AL. Designer Oncolytic Adenovirus: Coming of Age. Cancers (Basel) 2018; 10:E201. [PMID: 29904022 PMCID: PMC6025169 DOI: 10.3390/cancers10060201] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
The licensing of talimogene laherparepvec (T-Vec) represented a landmark moment for oncolytic virotherapy, since it provided unequivocal evidence for the long-touted potential of genetically modified replicating viruses as anti-cancer agents. Whilst T-Vec is promising as a locally delivered virotherapy, especially in combination with immune-checkpoint inhibitors, the quest continues for a virus capable of specific tumour cell killing via systemic administration. One candidate is oncolytic adenovirus (Ad); it’s double stranded DNA genome is easily manipulated and a wide range of strategies and technologies have been employed to empower the vector with improved pharmacokinetics and tumour targeting ability. As well characterised clinical and experimental agents, we have detailed knowledge of adenoviruses’ mechanisms of pathogenicity, supported by detailed virological studies and in vivo interactions. In this review we highlight the strides made in the engineering of bespoke adenoviral vectors to specifically infect, replicate within, and destroy tumour cells. We discuss how mutations in genes regulating adenoviral replication after cell entry can be used to restrict replication to the tumour, and summarise how detailed knowledge of viral capsid interactions enable rational modification to eliminate native tropisms, and simultaneously promote active uptake by cancerous tissues. We argue that these designer-viruses, exploiting the viruses natural mechanisms and regulated at every level of replication, represent the ideal platforms for local overexpression of therapeutic transgenes such as immunomodulatory agents. Where T-Vec has paved the way, Ad-based vectors now follow. The era of designer oncolytic virotherapies looks decidedly as though it will soon become a reality.
Collapse
Affiliation(s)
- Alexander T Baker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Carmen Aguirre-Hernández
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| |
Collapse
|
32
|
Lei J, Jacobus EJ, Taverner WK, Fisher KD, Hemmi S, West K, Slater L, Lilley F, Brown A, Champion B, Duffy MR, Seymour LW. Expression of human CD46 and trans-complementation by murine adenovirus 1 fails to allow productive infection by a group B oncolytic adenovirus in murine cancer cells. J Immunother Cancer 2018; 6:55. [PMID: 29898782 PMCID: PMC6000980 DOI: 10.1186/s40425-018-0350-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/07/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Oncolytic viruses are currently experiencing accelerated development in several laboratories worldwide, with some forty-seven clinical trials currently recruiting. Many oncolytic viruses combine targeted cytotoxicity to cancer cells with a proinflammatory cell lysis. Due to their additional potential to express immunomodulatory transgenes, they are also often known as oncolytic viral vaccines. However, several types of oncolytic viruses are human-specific and the lack of suitable immune-competent animal models complicates biologically relevant evaluation of their vaccine potential. This is a particular challenge for group B adenoviruses, which fail to infect even those immunocompetent animal model systems identified as semi-permissive for type 5 adenovirus. Here, we aim to develop a murine cell line capable of supporting replication of a group B oncolytic adenovirus, enadenotucirev (EnAd), for incorporation into a syngeneic immunocompetent animal model to explore the oncolytic vaccine potential of group B oncolytic viruses. METHODS Transgenic murine cell lines were infected with EnAd expressing GFP transgene under replication-independent or -dependent promoters. Virus mRNA expression, genome replication, and late protein expression were determined by qRT-PCR, qPCR, and immunoblotting, respectively. We also use Balb/c immune-competent mice to determine the tumourogenicity and infectivity of transgenic murine cell lines. RESULTS Our results show that a broad range of human carcinoma cells will support EnAd replication, but not murine carcinoma cells. Murine cells can be readily modified to express surface human CD46, one of the receptors for group B adenoviruses, allowing receptor-mediated uptake of EnAd particles into the murine cells and expression of CMV promoter-driven transgenes. Although the early E1A mRNA was expressed in murine cells at levels similar to human cells, adenovirus E2B and Fibre mRNA expression levels were hampered and few virus genomes were produced. Unlike previous reports on group C adenoviruses, trans-complementation of group B adenoviruses by co-infection with mouse adenovirus 1 did not rescue replication. A panel of group B adenoviruses expressing individual mouse adenovirus 1 genes were also unable to rescue EnAd replication. CONCLUSION Together, these results indicate that there may be major differences in the early stages of replication of group C and B adenoviruses in murine cells, and that the block to the life cycle of B adenoviruses in murine cells occurs in the early stage of virus replication, perhaps reflecting poor activity of Ad11p E1A in murine cells.
Collapse
Affiliation(s)
- Janet Lei
- 0000 0004 1936 8948grid.4991.5Department of OncologyUniversity of Oxford OX3 7DQ Oxford UK
| | - Egon J Jacobus
- 0000 0004 1936 8948grid.4991.5Department of OncologyUniversity of Oxford OX3 7DQ Oxford UK
| | - William K Taverner
- 0000 0004 1936 8948grid.4991.5Department of OncologyUniversity of Oxford OX3 7DQ Oxford UK
| | - Kerry D Fisher
- 0000 0004 1936 8948grid.4991.5Department of OncologyUniversity of Oxford OX3 7DQ Oxford UK
| | - Silvio Hemmi
- 0000 0004 1937 0650grid.7400.3Institute of Molecular Life SciencesUniversity of Zurich Zurich Switzerland
| | - Katy West
- 0000 0004 0394 8673grid.476643.4PsiOxus Therapeutics Ltd PsiOxus House, 4-10 The Quadrant, Barton Lane OX14 3YS Abingdon Oxfordshire UK
| | - Lorna Slater
- 0000 0004 0394 8673grid.476643.4PsiOxus Therapeutics Ltd PsiOxus House, 4-10 The Quadrant, Barton Lane OX14 3YS Abingdon Oxfordshire UK
| | - Fred Lilley
- 0000 0004 0394 8673grid.476643.4PsiOxus Therapeutics Ltd PsiOxus House, 4-10 The Quadrant, Barton Lane OX14 3YS Abingdon Oxfordshire UK
| | - Alice Brown
- 0000 0004 0394 8673grid.476643.4PsiOxus Therapeutics Ltd PsiOxus House, 4-10 The Quadrant, Barton Lane OX14 3YS Abingdon Oxfordshire UK
| | - Brian Champion
- 0000 0004 0394 8673grid.476643.4PsiOxus Therapeutics Ltd PsiOxus House, 4-10 The Quadrant, Barton Lane OX14 3YS Abingdon Oxfordshire UK
| | - Margaret R Duffy
- 0000 0004 1936 8948grid.4991.5Department of OncologyUniversity of Oxford OX3 7DQ Oxford UK
| | - Len W Seymour
- 0000 0004 1936 8948grid.4991.5Department of OncologyUniversity of Oxford OX3 7DQ Oxford UK
| |
Collapse
|
33
|
Reactivation of γ-globin in adult β-YAC mice after ex vivo and in vivo hematopoietic stem cell genome editing. Blood 2018; 131:2915-2928. [PMID: 29789357 DOI: 10.1182/blood-2018-03-838540] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
Abstract
Disorders involving β-globin gene mutations, primarily β-thalassemia and sickle cell disease, represent a major target for hematopoietic stem/progenitor cell (HSPC) gene therapy. This includes CRISPR/Cas9-mediated genome editing approaches in adult CD34+ cells aimed toward the reactivation of fetal γ-globin expression in red blood cells. Because models involving erythroid differentiation of CD34+ cells have limitations in assessing γ-globin reactivation, we focused on human β-globin locus-transgenic (β-YAC) mice. We used a helper-dependent human CD46-targeting adenovirus vector expressing CRISPR/Cas9 (HDAd-HBG-CRISPR) to disrupt a repressor binding region within the γ-globin promoter. We transduced HSPCs from β-YAC/human CD46-transgenic mice ex vivo and subsequently transplanted them into irradiated recipients. Furthermore, we used an in vivo HSPC transduction approach that involves HSPC mobilization and the intravenous injection of HDAd-HBG-CRISPR into β-YAC/CD46-transgenic mice. In both models, we demonstrated efficient target site disruption, resulting in a pronounced switch from human β- to γ-globin expression in red blood cells of adult mice that was maintained after secondary transplantation of HSPCs. In long-term follow-up studies, we did not detect hematological abnormalities, indicating that HBG promoter editing does not negatively affect hematopoiesis. This is the first study that shows successful in vivo HSPC genome editing by CRISPR/Cas9.
Collapse
|
34
|
Lundstrom K. Viral Vectors in Gene Therapy. Diseases 2018; 6:diseases6020042. [PMID: 29883422 PMCID: PMC6023384 DOI: 10.3390/diseases6020042] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 01/02/2023] Open
Abstract
Applications of viral vectors have found an encouraging new beginning in gene therapy in recent years. Significant improvements in vector engineering, delivery, and safety have placed viral vector-based therapy at the forefront of modern medicine. Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious diseases and different types of cancer. Recent development in the area of immunotherapy has provided both preventive and therapeutic approaches. Furthermore, gene silencing generating a reversible effect has become an interesting alternative, and is well-suited for delivery by viral vectors. A number of preclinical studies have demonstrated therapeutic and prophylactic efficacy in animal models and furthermore in clinical trials. Several viral vector-based drugs have also been globally approved.
Collapse
|
35
|
Duffy MR, Fisher KD, Seymour LW. Making Oncolytic Virotherapy a Clinical Reality: The European Contribution. Hum Gene Ther 2017; 28:1033-1046. [PMID: 28793793 DOI: 10.1089/hum.2017.112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Oncolytic viruses (OVs) are quickly moving toward the forefront of modern medicines. The reward for the decades of research invested into developing viral platforms that selectively replicate in and lyse tumor cells while sparking anticancer adaptive immunity is presenting in the form of durable therapeutic responses. While this has certainly been a concerted global effort, in this review for the 25th anniversary of the European Society of Gene and Cell Therapy, we focus on the contributions made by European researchers. Research centers across Europe have held central roles in advancing OVs, from the earliest reports of coincidental viral infections leading to antitumor efficacy, to advanced mechanistic studies, and now through Phase I-III trials to imminent regulatory approvals. While challenges still remain, with limitations in preclinical animal models, antiviral immune clearance, and manufacture restrictions enforced by poor viral yields in certain cases, the field has come a very long way in recent years. Thoughtful mechanistic integration of OVs with standard of care strategies and other newly approved therapies should provide potent novel approaches. Combination with immunotherapeutic regimes holds significant promise, and the ability to arm the viral platform with therapeutic proteins for localized expression at the tumor site provides an opportunity for creating highly effective synergistic treatments and brings a new age of targeted cancer therapeutics.
Collapse
Affiliation(s)
- Margaret R Duffy
- Department of Oncology, University of Oxford , Oxford, United Kingdom
| | - Kerry D Fisher
- Department of Oncology, University of Oxford , Oxford, United Kingdom
| | - Len W Seymour
- Department of Oncology, University of Oxford , Oxford, United Kingdom
| |
Collapse
|
36
|
Garcia-Carbonero R, Salazar R, Duran I, Osman-Garcia I, Paz-Ares L, Bozada JM, Boni V, Blanc C, Seymour L, Beadle J, Alvis S, Champion B, Calvo E, Fisher K. Phase 1 study of intravenous administration of the chimeric adenovirus enadenotucirev in patients undergoing primary tumor resection. J Immunother Cancer 2017; 5:71. [PMID: 28923104 PMCID: PMC5604344 DOI: 10.1186/s40425-017-0277-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/14/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Enadenotucirev (formerly ColoAd1) is a tumor-selective chimeric adenovirus with demonstrated preclinical activity. This phase 1 Mechanism of Action study assessed intravenous (IV) delivery of enadenotucirev in patients with resectable colorectal cancer (CRC), non-small-cell lung cancer (NSCLC), urothelial cell cancer (UCC), and renal cell cancer (RCC) with a comparator intratumoral (IT) dosed CRC patient cohort. METHODS Seventeen patients scheduled for primary tumor resection were enrolled. IT injection of enadenotucirev (CRC only) was administered as a single dose (≤ 3 × 1011 viral particles [vp]) on day 1, followed by resection during days 8-15. IV infusion of enadenotucirev was administered by three separate doses (1 × 1012 vp) on days 1, 3, and 5, followed by resection during days 8-15 (CRC) or days 10-25 (NSCLC, UCC, and RCC). Enadenotucirev activity was measured using immunohistochemical staining of nuclear viral hexon and quantitative polymerase chain reaction for viral genomic DNA. RESULTS Delivery of enadenotucirev was observed in most tumor samples following IV infusion, with little or no demonstrable activity in normal tissue. This virus delivery (by both IV and IT dosing) was accompanied by high local CD8+ cell infiltration in 80% of tested tumor samples, suggesting a potential enadenotucirev-driven immune response. Both methods of enadenotucirev delivery were well tolerated, with no treatment-associated serious adverse events. CONCLUSIONS This study provides key delivery and feasibility data to support the use of IV infusion of enadenotucirev, or therapeutic transgene-bearing derivatives of it, in clinical trials across a range of epithelial tumors, including the ongoing combination study of enadenotucirev with the checkpoint inhibitor nivolumab. It also provides insights into the potential immune-stimulating properties of enadenotucirev. TRIAL REGISTRATION This MOA study was a phase 1, multicenter, non-randomized, open-label study to investigate the administration of enadenotucirev in a preoperative setting (ClinicalTrials.gov: NCT02053220).
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Adenoviruses, Human/physiology
- Administration, Intravenous
- CD8-Positive T-Lymphocytes/metabolism
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/therapy
- Carcinoma, Transitional Cell/immunology
- Carcinoma, Transitional Cell/therapy
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/therapy
- Combined Modality Therapy
- DNA, Viral/genetics
- Digestive System Surgical Procedures
- Humans
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Oncolytic Virotherapy
- Oncolytic Viruses/genetics
- Oncolytic Viruses/physiology
- Pulmonary Surgical Procedures
- Treatment Outcome
- Urologic Surgical Procedures
Collapse
Affiliation(s)
- Rocio Garcia-Carbonero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Ramon Salazar
- Medical Oncology Department, Catalan Institute of Oncology, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Ignacio Duran
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Ignacio Osman-Garcia
- Unidad de Urología-Oncológica, UGC de Urología y Nefrología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidadde Sevilla, Seville, Spain
| | - Luis Paz-Ares
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Juan M. Bozada
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Valentina Boni
- START Madrid, Centro Integral Oncológico Clara Campal, Hospital Madrid Norte Sanchinarro, Madrid, Spain
| | | | - Len Seymour
- Department of Oncology, Oxford University, Oxford, UK
| | - John Beadle
- PsiOxus Therapeutics Limited, Milton Park, Abingdon, UK
| | - Simon Alvis
- PsiOxus Therapeutics Limited, Milton Park, Abingdon, UK
| | | | - Emiliano Calvo
- START Madrid, Centro Integral Oncológico Clara Campal, Hospital Madrid Norte Sanchinarro, Madrid, Spain
| | - Kerry Fisher
- PsiOxus Therapeutics Limited, Milton Park, Abingdon, UK
- Department of Oncology, Oxford University, Oxford, UK
| |
Collapse
|
37
|
Freedman JD, Hagel J, Scott EM, Psallidas I, Gupta A, Spiers L, Miller P, Kanellakis N, Ashfield R, Fisher KD, Duffy MR, Seymour LW. Oncolytic adenovirus expressing bispecific antibody targets T-cell cytotoxicity in cancer biopsies. EMBO Mol Med 2017; 9:1067-1087. [PMID: 28634161 PMCID: PMC5538299 DOI: 10.15252/emmm.201707567] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 12/21/2022] Open
Abstract
Oncolytic viruses exploit the cancer cell phenotype to complete their lytic life cycle, releasing progeny virus to infect nearby cells and repeat the process. We modified the oncolytic group B adenovirus EnAdenotucirev (EnAd) to express a bispecific single-chain antibody, secreted from infected tumour cells into the microenvironment. This bispecific T-cell engager (BiTE) binds to EpCAM on target cells and cross-links them to CD3 on T cells, leading to clustering and activation of both CD4 and CD8 T cells. BiTE transcription can be controlled by the virus major late promoter, limiting expression to cancer cells that are permissive for virus replication. This approach can potentiate the cytotoxicity of EnAd, and we demonstrate using primary pleural effusions and peritoneal malignant ascites that infection of cancer cells with the BiTE-expressing EnAd leads to activation of endogenous T cells to kill endogenous tumour cells despite the immunosuppressive environment. In this way, we have armed EnAd to combine both direct oncolysis and T cell-mediated killing, yielding a potent therapeutic that should be readily transferred into the clinic.
Collapse
Affiliation(s)
| | - Joachim Hagel
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Ioannis Psallidas
- Oxford Centre for Respiratory Medicine, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Avinash Gupta
- Churchill Hospital, Oxford University Hospital NHS Trust, Oxford, UK
| | - Laura Spiers
- Churchill Hospital, Oxford University Hospital NHS Trust, Oxford, UK
| | - Paul Miller
- Churchill Hospital, Oxford University Hospital NHS Trust, Oxford, UK
| | - Nikolaos Kanellakis
- Oxford Centre for Respiratory Medicine, Oxford University Hospitals NHS Trust, Oxford, UK
| | | | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
38
|
|