1
|
Wallace R, Bliss CM, Parker AL. The Immune System-A Double-Edged Sword for Adenovirus-Based Therapies. Viruses 2024; 16:973. [PMID: 38932265 PMCID: PMC11209478 DOI: 10.3390/v16060973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Pathogenic adenovirus (Ad) infections are widespread but typically mild and transient, except in the immunocompromised. As vectors for gene therapy, vaccine, and oncology applications, Ad-based platforms offer advantages, including ease of genetic manipulation, scale of production, and well-established safety profiles, making them attractive tools for therapeutic development. However, the immune system often poses a significant challenge that must be overcome for adenovirus-based therapies to be truly efficacious. Both pre-existing anti-Ad immunity in the population as well as the rapid development of an immune response against engineered adenoviral vectors can have detrimental effects on the downstream impact of an adenovirus-based therapeutic. This review focuses on the different challenges posed, including pre-existing natural immunity and anti-vector immunity induced by a therapeutic, in the context of innate and adaptive immune responses. We summarise different approaches developed with the aim of tackling these problems, as well as their outcomes and potential future applications.
Collapse
Affiliation(s)
- Rebecca Wallace
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
| | - Carly M. Bliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alan L. Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
2
|
Cervantes-Torres J, Cabello-Gutiérrez C, Ayón-Núñez DA, Soldevila G, Olguin-Alor R, Diaz G, Acero G, Segura-Velázquez R, Huerta L, Gracia-Mora I, Cobos L, Pérez-Tapia M, Almagro JC, Suárez-Güemes F, Bobes RJ, Fragoso G, Sciutto E, Laclette JP. Caveats of chimpanzee ChAdOx1 adenovirus-vectored vaccines to boost anti-SARS-CoV-2 protective immunity in mice. Appl Microbiol Biotechnol 2024; 108:179. [PMID: 38280035 PMCID: PMC10821985 DOI: 10.1007/s00253-023-12927-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/21/2023] [Accepted: 11/26/2023] [Indexed: 01/29/2024]
Abstract
Several COVID-19 vaccines use adenovirus vectors to deliver the SARS-CoV-2 spike (S) protein. Immunization with these vaccines promotes immunity against the S protein, but against also the adenovirus itself. This could interfere with the entry of the vaccine into the cell, reducing its efficacy. Herein, we evaluate the efficiency of an adenovirus-vectored vaccine (chimpanzee ChAdOx1 adenovirus, AZD1222) in boosting the specific immunity compared to that induced by a recombinant receptor-binding domain (RBD)-based vaccine without viral vector. Mice immunized with the AZD1222 human vaccine were given a booster 6 months later, with either the homologous vaccine or a recombinant vaccine based on RBD of the delta variant, which was prevalent at the start of this study. A significant increase in anti-RBD antibody levels was observed in rRBD-boosted mice (31-61%) compared to those receiving two doses of AZD1222 (0%). Significantly higher rates of PepMix™- or RBD-elicited proliferation were also observed in IFNγ-producing CD4 and CD8 cells from mice boosted with one or two doses of RBD, respectively. The lower efficiency of the ChAdOx1-S vaccine in boosting specific immunity could be the result of a pre-existing anti-vector immunity, induced by increased levels of anti-adenovirus antibodies found both in mice and humans. Taken together, these results point to the importance of avoiding the recurrent use of the same adenovirus vector in individuals with immunity and memory against them. It also illustrates the disadvantages of ChAdOx1 adenovirus-vectored vaccine with respect to recombinant protein vaccines, which can be used without restriction in vaccine-booster programs. KEY POINTS: • ChAdOx1 adenovirus vaccine (AZD1222) may not be effective in boosting anti-SARS-CoV-2 immunity • A recombinant RBD protein vaccine is effective in boosting anti-SARS-CoV-2 immunity in mice • Antibodies elicited by the rRBD-delta vaccine persisted for up to 3 months in mice.
Collapse
Affiliation(s)
- Jacquelynne Cervantes-Torres
- School of Veterinary Medicine, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Carlos Cabello-Gutiérrez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Belisario Domínguez Secc. 16, Tlalpan, 14080, Mexico City, CDMX, Mexico
| | - Dolores-Adriana Ayón-Núñez
- School of Veterinary Medicine, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Gloria Soldevila
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Roxana Olguin-Alor
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Georgina Diaz
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Gonzalo Acero
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - René Segura-Velázquez
- School of Veterinary Medicine, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Leonor Huerta
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Isabel Gracia-Mora
- Unidad de Experimentación Preclínica, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Laura Cobos
- School of Veterinary Medicine, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapeúticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, 11340, Mexico City, Mexico
| | - Juan C Almagro
- Unidad de Desarrollo e Investigación en Bioterapeúticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, 11340, Mexico City, Mexico
| | - Francisco Suárez-Güemes
- School of Veterinary Medicine, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Raúl J Bobes
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Gladis Fragoso
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico
| | - Edda Sciutto
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico.
| | - Juan Pedro Laclette
- Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
3
|
Wang X, Hetzel M, Zhang W, Ehrhardt A, Bayer W. Comparative analysis of the impact of 40 adenovirus types on dendritic cell activation and CD8 + T cell proliferation capacity for the identification of favorable immunization vector candidates. Front Immunol 2023; 14:1286622. [PMID: 37915567 PMCID: PMC10616870 DOI: 10.3389/fimmu.2023.1286622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
For the development of new adenovirus (AdV)-based vectors, it is important to understand differences in immunogenicity. In a side-by-side in vitro analysis, we evaluated the effect of 40 AdV types covering human AdV (HAdV) species A through G on the expression of 11 activation markers and the secretion of 12 cytokines by AdV-transduced dendritic cells, and the effect on CD8+ T cell proliferation capacity. We found that the expression of activation markers and cytokines differed widely between the different HAdV types, and many types were able to significantly impair the proliferation capacity of CD8+ T cells. Univariate and multivariate regression analyses suggested an important role of type I interferons in mediating this suppression of CD8+ T cells, which we confirmed experimentally in a proliferation assay using a type I interferon receptor blocking antibody. Using Bayesian statistics, we calculated a prediction model that suggests HAdV types HAdV-C1, -D8, -B7, -F41, -D33, -C2, -A31, -B3 and -D65 as the most favorable candidates for vaccine vector development.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mario Hetzel
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
4
|
Paris O, Mennechet FJD, Kremer EJ. Human innate lymphoid cell activation by adenoviruses is modified by host defense proteins and neutralizing antibodies. Front Immunol 2022; 13:975910. [PMID: 36275713 PMCID: PMC9579290 DOI: 10.3389/fimmu.2022.975910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Innate lymphoid cells (ILCs), the complements of diverse CD4 T helper cells, help maintain tissue homeostasis by providing a link between innate and adaptive immune responses. While pioneering studies over the last decade have advanced our understanding how ILCs influence adaptive immune responses to pathogens, far less is known about whether the adaptive immune response feeds back into an ILC response. In this study, we isolated ILCs from blood of healthy donors, fine-tuned culture conditions, and then directly challenged them with human adenoviruses (HAdVs), with HAdVs and host defense proteins (HDPs) or neutralizing antibodies (NAbs), to mimic interactions in a host with pre-existing immunity. Additionally, we developed an ex vivo approach to identify how bystander ILCs respond to the uptake of HAdVs ± neutralizing antibodies by monocyte-derived dendritic cells. We show that ILCs take up HAdVs, which induces phenotypic maturation and cytokine secretion. Moreover, NAbs and HDPs complexes modified the cytokine profile generated by ILCs, consistent with a feedback loop for host antiviral responses and potential to impact adenovirus-based vaccine efficacy.
Collapse
|
5
|
Dienst EGT, Kremer EJ. Adenovirus receptors on antigen-presenting cells of the skin. Biol Cell 2022; 114:297-308. [PMID: 35906865 DOI: 10.1111/boc.202200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 12/01/2022]
Abstract
Skin, the largest human organ, is part of the first line of physical and immunological defense against many pathogens. Understanding how skin antigen-presenting cells (APCs) respond to viruses or virus-based vaccines is crucial to develop antiviral pharmaceutics, and efficient and safe vaccines. Here, we discuss the way resident and recruited skin APCs engage adenoviruses and the impact on innate immune responses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
6
|
Keshavarz M, Mohammad Miri S, Behboudi E, Arjeini Y, Dianat-Moghadam H, Ghaemi A. Oncolytic virus delivery modulated immune responses toward cancer therapy: Challenges and perspectives. Int Immunopharmacol 2022; 108:108882. [PMID: 35623296 DOI: 10.1016/j.intimp.2022.108882] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 11/05/2022]
Abstract
Oncolytic viruses (OVs) harness the hallmarks of tumor cells and cancer-related immune responses for the lysis of malignant cells, modulation of the tumor microenvironment, and exertion of vaccine-like activities. However, efficient clinical exploitation of these potent therapeutic modules requires their systematic administration, especially against metastatic and solid tumors. Therefore, developing methods for shielding a virus from the neutralizing environment of the bloodstream while departing toward tumor sites is a must. This paper reports the latest advancements in the employment of chemical and biological compounds aimed at safe and efficient delivery of OVs to target tissues or tumor deposits within the host.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Seyed Mohammad Miri
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| | - Emad Behboudi
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Yaser Arjeini
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran.
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
Cytokine Responses to Adenovirus and Adenovirus Vectors. Viruses 2022; 14:v14050888. [PMID: 35632630 PMCID: PMC9145601 DOI: 10.3390/v14050888] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
The expression of cytokines and chemokines in response to adenovirus infection is tightly regulated by the innate immune system. Cytokine-mediated toxicity and cytokine storm are known clinical phenomena observed following naturally disseminated adenovirus infection in immunocompromised hosts as well as when extremely high doses of adenovirus vectors are injected intravenously. This dose-dependent, cytokine-mediated toxicity compromises the safety of adenovirus-based vectors and represents a critical problem, limiting their utility for gene therapy applications and the therapy of disseminated cancer, where intravenous injection of adenovirus vectors may provide therapeutic benefits. The mechanisms triggering severe cytokine response are not sufficiently understood, prompting efforts to further investigate this phenomenon, especially in clinically relevant settings. In this review, we summarize the current knowledge on cytokine and chemokine activation in response to adenovirus- and adenovirus-based vectors and discuss the underlying mechanisms that may trigger acute cytokine storm syndrome. First, we review profiles of cytokines and chemokines that are activated in response to adenovirus infection initiated via different routes. Second, we discuss the molecular mechanisms that lead to cytokine and chemokine transcriptional activation. We further highlight how immune cell types in different organs contribute to synthesis and systemic release of cytokines and chemokines in response to adenovirus sensing. Finally, we review host factors that can limit cytokine and chemokine expression and discuss currently available and potential future interventional approaches that allow for the mitigation of the severity of the cytokine storm syndrome. Effective cytokine-targeted interventional approaches may improve the safety of systemic adenovirus delivery and thus broaden the potential clinical utility of adenovirus-based therapeutic vectors.
Collapse
|
8
|
Elkoshi Z. SARS-CoV-2 Omicron (B.1.1.529) Variant: Corticosteroids Treatment/Respiratory Coinfection. Front Immunol 2022; 13:856072. [PMID: 35309339 PMCID: PMC8927656 DOI: 10.3389/fimmu.2022.856072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/10/2022] [Indexed: 02/05/2023] Open
Affiliation(s)
- Zeev Elkoshi
- Research and Development Department, Taro Pharmaceutical Industries Ltd., Haifa, Israel
| |
Collapse
|
9
|
Adenovirus-α-defensin complexes induce NLRP3-associated maturation of human phagocytes via TLR4 engagement. J Virol 2022; 96:e0185021. [PMID: 35080426 DOI: 10.1128/jvi.01850-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intramuscular delivery of human adenovirus (HAdV)-based vaccines leads to rapid recruitment of neutrophils, which then release antimicrobial peptides/proteins (AMPs). How these AMPs influence vaccine efficacy over the subsequent 24 h is poorly understood. In this study, we asked if human neutrophil protein 1 (HNP-1), an α-defensin that influences the direct and indirect innate immune responses to a range of pathogens, impacts the response of human phagocytes to three HAdV species/types (HAdV-C5, -D26, -B35). We show that HNP-1 binds to the capsids, redirects HAdV-C5, -D26, -B35 to Toll-like receptor 4 (TLR4), which leads to internalization, an NLRP3-mediated inflammasome response, and IL-1β release. Surprisingly, IL-1β release was not associated with notable disruption of plasma membrane integrity. These data further our understanding of HAdV vaccine immunogenicity and may provide pathways to extend the efficacy. Importance This study examines the interactions between danger-associated molecular patterns and human adenoviruses and its impact on vaccines. HAdVs and HNP-1 can interact, these interactions will modify the response of antigen-presenting cells., which will influence vaccine efficacy.
Collapse
|
10
|
Tran TTP, Tran TH, Kremer EJ. IgG-Complexed Adenoviruses Induce Human Plasmacytoid Dendritic Cell Activation and Apoptosis. Viruses 2021; 13:1699. [PMID: 34578281 PMCID: PMC8472521 DOI: 10.3390/v13091699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Following repeat exposure to many human adenoviruses (HAdVs), most adults harbour long-lived B- and T-cell responses. Combined, this response typically protects us for years from re-infection by the same HAdV type. In spite of these immune responses, some HAdV types are associated with persistent infections that constitute a life-threatening risk when an individual's T-cell response is compromised. By contrast, patients with B-cell deficiencies do not appear to be at a greater risk of HAdV disease. This dichotomy begs the question of the secondary role of anti-HAdV antibodies during host defence. In this study, we explored IgG-complexed (IC)-HAdV5 and primary human plasmacytoid dendritic cell (pDC) interactions. We found that IC-HAdV5 are efficiently internalized in pDCs, stimulate their activation through TLR9 signalling, and cause apoptosis. These data may help reconcile the enigma of robust immune response to HAdVs, while concurrently allowing persistence.
Collapse
Affiliation(s)
- Thi Thu Phuong Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, 34090 Montpellier, France; (T.T.P.T.); (T.H.T.)
- Department of Life Sciences, University of Science and Technology of Hanoi Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
| | - Tuan Hiep Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, 34090 Montpellier, France; (T.T.P.T.); (T.H.T.)
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
- PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, Hanoi 11313, Vietnam
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, 34090 Montpellier, France; (T.T.P.T.); (T.H.T.)
| |
Collapse
|
11
|
Obrová K, Grumaz S, Remely M, Czurda S, Krickl I, Herndlhofer S, Gleixner KV, Sperr WR, Größlinger L, Frank T, Andrade N, Egger‐Matiqi T, Peters C, Engstler G, Dworzak M, Attarbaschi A, Grotel M, Heuvel‐Eibrink MM, Moiseev IS, Rogacheva Y, Zubarovskaya L, Zubarovskaya N, Pichler H, Lawitschka A, Koller E, Keil F, Valent P, Sohn K, Lion T. Presence of viremia during febrile neutropenic episodes in patients undergoing chemotherapy for malignant neoplasms. Am J Hematol 2021; 96:719-726. [PMID: 33784434 DOI: 10.1002/ajh.26177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022]
Abstract
The importance of viral infections as a leading cause of morbidity and mortality is well documented in severely immunosuppressed patients undergoing allogeneic stem cell transplantation. By contrast, viral infections generally receive less attention in patients with malignant disorders undergoing chemotherapy, where the onset of neutropenic fever is mostly associated with bacterial or fungal infections, and screening for viral infections is not routinely performed. To address the occurrence of invasive viral infections in a clinical setting commonly associated with less pronounced immunosuppression, we have prospectively screened 237 febrile neutropenic episodes in pediatric (n = 77) and adult (n = 69) patients undergoing intensive chemotherapy, primarily for treatment of acute leukemia. Serial peripheral blood specimens were tested by RQ-PCR assays for the presence and quantity of the clinically relevant viruses CMV, EBV, HHV6 and HAdV, commonly reactivated in highly immunocompromised patients. Viremia was documented in 36 (15%) episodes investigated, including the detection of HHV6 (n = 14), EBV (n = 15), CMV (n = 6), or HAdV (n = 1). While low or intermediate levels of viremia (<104 virus copies/mL) were commonly associated with bacterial or fungal co-infection, viremia at higher levels (>104 copies/mL) was documented in patients without evidence for other infections, raising the possibility that at least in some instances the onset of fever may have been attributable to the virus detected. The observations suggest that viral infections, potentially resulting from reactivation, might also play a clinically relevant role in patients receiving chemotherapy for treatment of malignant neoplasms, and routine screening for viremia in this clinical setting might be warranted.
Collapse
Affiliation(s)
- Klára Obrová
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
| | | | - Marlene Remely
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
| | - Stefan Czurda
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
| | - Isabella Krickl
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
| | - Susanne Herndlhofer
- Department of Internal Medicine I, Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
| | - Karoline V. Gleixner
- Department of Internal Medicine I, Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Institute for Hematology & Oncology Medical University of Vienna Vienna Austria
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Institute for Hematology & Oncology Medical University of Vienna Vienna Austria
| | | | - Tijana Frank
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
| | - Nuno Andrade
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
| | - Teresa Egger‐Matiqi
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Christina Peters
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Gernot Engstler
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Michael Dworzak
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Andishe Attarbaschi
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Martine Grotel
- Princess Máxima Center for Pediatric Oncology Utrecht The Netherlands
| | | | - Ivan S. Moiseev
- I. P. Pavlov First Saint Petersburg State Medical University Saint Petersburg Russia
| | - Yuliya Rogacheva
- I. P. Pavlov First Saint Petersburg State Medical University Saint Petersburg Russia
| | - Ludmilla Zubarovskaya
- I. P. Pavlov First Saint Petersburg State Medical University Saint Petersburg Russia
| | - Natalia Zubarovskaya
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Herbert Pichler
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | - Anita Lawitschka
- St. Anna Children's Hospital, Department of Pediatrics Medical University of Vienna Vienna Austria
| | | | - Felix Keil
- Ludwig Boltzmann Institute for Hematology & Oncology Medical University of Vienna Vienna Austria
- 3rd Medical Department Hanuschhospital Vienna Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology Medical University of Vienna Vienna Austria
- Ludwig Boltzmann Institute for Hematology & Oncology Medical University of Vienna Vienna Austria
| | - Kai Sohn
- Fraunhofer IGB Stuttgart Germany
| | - Thomas Lion
- St.Anna Children's Cancer Research Institute (CCRI) Vienna Austria
- Ludwig Boltzmann Institute for Hematology & Oncology Medical University of Vienna Vienna Austria
- Labdia Labordiagnostik GmbH Vienna Austria
- Department of Pediatrics Medical University of Vienna Vienna Austria
| |
Collapse
|
12
|
Chéneau C, Eichholz K, Tran TH, Tran TTP, Paris O, Henriquet C, Bajramovic JJ, Pugniere M, Kremer EJ. Lactoferrin Retargets Human Adenoviruses to TLR4 to Induce an Abortive NLRP3-Associated Pyroptotic Response in Human Phagocytes. Front Immunol 2021; 12:685218. [PMID: 34093588 PMCID: PMC8173049 DOI: 10.3389/fimmu.2021.685218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
Despite decades of clinical and preclinical investigations, we still poorly grasp our innate immune response to human adenoviruses (HAdVs) and their vectors. In this study, we explored the impact of lactoferrin on three HAdV types that are being used as vectors for vaccines. Lactoferrin is a secreted globular glycoprotein that influences direct and indirect innate immune response against a range of pathogens following a breach in tissue homeostasis. The mechanism by which lactoferrin complexes increases HAdV uptake and induce maturation of human phagocytes is unknown. We show that lactoferrin redirects HAdV types from species B, C, and D to Toll-like receptor 4 (TLR4) cell surface complexes. TLR4-mediated internalization of the HAdV-lactoferrin complex induced an NLRP3-associated response that consisted of cytokine release and transient disruption of plasma membrane integrity, without causing cell death. These data impact our understanding of HAdV immunogenicity and may provide ways to increase the efficacy of HAdV-based vectors/vaccines.
Collapse
Affiliation(s)
- Coraline Chéneau
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Karsten Eichholz
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Tuan Hiep Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Thi Thu Phuong Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Océane Paris
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Corinne Henriquet
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université Montpellier, Institut Régional du Cancer, Montpellier, France
| | | | - Martine Pugniere
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université Montpellier, Institut Régional du Cancer, Montpellier, France
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
13
|
Shin DH, Nguyen T, Ozpolat B, Lang F, Alonso M, Gomez-Manzano C, Fueyo J. Current strategies to circumvent the antiviral immunity to optimize cancer virotherapy. J Immunother Cancer 2021; 9:jitc-2020-002086. [PMID: 33795384 PMCID: PMC8021759 DOI: 10.1136/jitc-2020-002086] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer virotherapy is a paradigm-shifting treatment modality based on virus-mediated oncolysis and subsequent antitumor immune responses. Clinical trials of currently available virotherapies showed that robust antitumor immunity characterizes the remarkable and long-term responses observed in a subset of patients. These data suggest that future therapies should incorporate strategies to maximize the immunotherapeutic potential of oncolytic viruses. In this review, we highlight the recent evidence that the antiviral immunity of the patients may limit the immunotherapeutic potential of oncolytic viruses and summarize the most relevant approaches to strategically redirect the immune response away from the viruses and toward tumors to heighten the clinical impact of viro-immunotherapy platforms.
Collapse
Affiliation(s)
- Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Teresa Nguyen
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederick Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marta Alonso
- Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
14
|
Elkoshi Z. The Binary Classification of Protein Kinases. J Inflamm Res 2021; 14:929-947. [PMID: 33776467 PMCID: PMC7988341 DOI: 10.2147/jir.s303750] [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: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
In an earlier publication a binary model for chronic diseases classification has been proposed. According to the model, chronic diseases were classified as “high Treg” or “low Treg” diseases, depending on whether the immune response is anti- or pro-inflammatory and assuming that regulatory T cells are major determinants of the response. It turned out that most cancers are “high Treg” diseases, while autoimmune diseases are “low Treg”. This paper proposes a molecular cause for this binary response. The mechanism proposed depends on the effect of protein kinases on the immune system. Thus, protein kinases are classified as anti- or pro-inflammatory kinases depending on whether they drive “high Treg” or “low Treg” diseases. Observations reported in the earlier publication can be described in terms of anti-inflammatory kinase (AIK) or pro-inflammatory kinase (PIK) activity. Analysis of literature data reveals that the two classes of kinases display distinctive properties relating to their interactions with pathogens and environmental factors. Pathogens that promote Treg activity (“high Treg” pathogens) activate AIKs, while pathogens that suppress Treg activity (“low Treg” pathogens) activate PIKs. Diseases driven by AIKs are associated with “high Treg” pathogens while those diseases driven by PIKs are associated with “low Treg” pathogens. By promoting the activity of AIKs, alcohol consumption increases the risk of “high Treg” cancers but decreases the risk of some “low Treg” autoimmune diseases. JAK1 gain-of-function mutations are observed at high frequencies in autoimmune diseases while JAK1 loss-of-function mutations are observed at high frequencies in cancers with high tumor-infiltrating Tregs. It should also be noted that the corresponding two classes of protein kinase inhibitors are mutually exclusive in terms of their approved therapeutic indications. There is no protein kinase inhibitor that is approved for the treatment of both autoimmune diseases and “high Treg” cancers. Although there are exceptions to the conclusions presented above, these conclusions are supported by the great bulk of published data. It therefore seems that the binary division of protein kinases is a useful tool for elucidating (at the molecular level) many distinctive properties of cancers and autoimmune diseases.
Collapse
Affiliation(s)
- Zeev Elkoshi
- Research and Development Department, Taro Pharmaceutical Industries Ltd, Haifa, Israel
| |
Collapse
|
15
|
Vupputuri S, Tayebi L, Hikkaduwa Koralege RS, Nigatu A, Mozafari M, Mishra A, Liu L, Ramsey JD. Polyethylene glycol–modified DOTAP:cholesterol/adenovirus hybrid vectors have improved transduction efficiency and reduced immunogenicity. JOURNAL OF NANOPARTICLE RESEARCH 2021; 23:37. [DOI: 10.1007/s11051-020-05134-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 12/26/2020] [Indexed: 03/07/2025]
|
16
|
Asha K, Khanna M, Kumar B. Current Insights into the Host Immune Response to Respiratory Viral Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:59-83. [PMID: 34661891 DOI: 10.1007/978-3-030-67452-6_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Respiratory viral infections often lead to severe illnesses varying from mild or asymptomatic upper respiratory tract infections to severe bronchiolitis and pneumonia or/and chronic obstructive pulmonary disease. Common viral infections, including but not limited to influenza virus, respiratory syncytial virus, rhinovirus and coronavirus, are often the leading cause of morbidity and mortality. Since the lungs are continuously exposed to foreign particles, including respiratory pathogens, it is also well equipped for recognition and antiviral defense utilizing the complex network of innate and adaptive immune cells. Immediately upon infection, a range of proinflammatory cytokines, chemokines and an interferon response is generated, thereby making the immune response a two edged sword, on one hand it is required to eliminate viral pathogens while on other hand it's prolonged response can lead to chronic infection and significant pulmonary damage. Since vaccines to all respiratory viruses are not available, a better understanding of the virus-host interactions, leading to the development of immune response, is critically needed to design effective therapies to limit the severity of inflammatory damage, enhance viral clearance and to compliment the current strategies targeting the virus. In this chapter, we discuss the host responses to common respiratory viral infections, the key players of adaptive and innate immunity and the fine balance that exists between the viral clearance and immune-mediated damage.
Collapse
Affiliation(s)
- Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Madhu Khanna
- Department of Virology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Binod Kumar
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
17
|
Dai S, Lv Y, Xu W, Yang Y, Liu C, Dong X, Zhang H, Prabhakar BS, Maker AV, Seth P, Wang H. Oncolytic adenovirus encoding LIGHT (TNFSF14) inhibits tumor growth via activating anti-tumor immune responses in 4T1 mouse mammary tumor model in immune competent syngeneic mice. Cancer Gene Ther 2020; 27:923-933. [PMID: 32307442 DOI: 10.1038/s41417-020-0173-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/17/2020] [Accepted: 03/26/2020] [Indexed: 12/24/2022]
Abstract
LIGHT, also known as tumor-necrosis factor (TNF) superfamily member 14 (TNFSF14), is predominantly expressed on activated immune cells and some tumor cells. LIGHT is a pivotal regulator both for recruiting and activating immune cells in the tumor lesions. In this study, we armed human telomerase reverse transcriptase (TERT) promoter controlled oncolytic adenovirus with LIGHT to generate rAd.Light. rAd.Light effectively transduced both human and mouse breast tumor cell lines in vitro, and expressed LIGHT protein on the surface of tumor cells. Both rAd.Null, and rAd.Light could replicate in human breast cancer cells, and produced cytotoxicity to human and mouse mammary tumor cells. rAd.Light induced apoptosis resulting in tumor cell death. Using a subcutaneous model of 4T1 cells in BALB/c mice, rAd.Light was delivered intratumorally to evaluate the anti-tumor responses. Both rAd.Light and rAd.Null significantly inhibited the tumor growth, but rAd.Light produced much stronger anti-tumor effects. Histopathological analysis showed the infiltration of T lymphocytes in the tumor tissues. rAd.Light also induced stronger cellular apoptosis than rAd.Null in the tumors. Interestingly, on day 15, compared to rAd.Null, there was a significant reduction of Tregs following rAd.Light treatment. rAd.Light significantly increased Th1 cytokine interleukin (IL)-2 expression, and reduced Th2 cytokines expression, such as transforming growth factor β (TGF-β) and IL-10 in the tumors. These results suggest rAd.Light induced activation of anti-tumor immune responses. In conclusion, rAd.Light produced anti-tumor effect in a subcutaneous model of breast cancer via inducing tumor apoptosis and evoking strong anti-tumor immune responses. Therefore, rAd.Light has great promise to be developed as an effective therapeutic approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shiyun Dai
- Anhui Medical University, Hefei, 230032, Anhui, PR China
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Yun Lv
- Anhui Medical University, Hefei, 230032, Anhui, PR China
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Weidong Xu
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, IL, USA
| | - Yuefeng Yang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, IL, USA
- Department of Experimental Medical Science & Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, 315000, Zhejiang, PR China
| | - Chao Liu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
- Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Xiwen Dong
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Huan Zhang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, PR China
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ajay V Maker
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Surgery, Division of Surgical Oncology, University of Illinois, Chicago, IL, USA
| | - Prem Seth
- Gene Therapy Program, Department of Medicine, NorthShore Research Institute, Evanston, IL, USA.
| | - Hua Wang
- Anhui Medical University, Hefei, 230032, Anhui, PR China.
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China.
| |
Collapse
|
18
|
Chéneau C, Kremer EJ. Adenovirus-Extracellular Protein Interactions and Their Impact on Innate Immune Responses by Human Mononuclear Phagocytes. Viruses 2020; 12:v12121351. [PMID: 33255892 PMCID: PMC7760109 DOI: 10.3390/v12121351] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
The aim of this review is to highlight how, in a syngeneic system, human mononuclear phagocytes respond to environments containing human adenovirus (HAdV) and soluble extracellular proteins that influence their innate immune response. Soluble extracellular proteins, including immunoglobulins, blood clotting factors, proteins of the complement system, and/or antimicrobial peptides (AMPs) can exert direct effects by binding to a virus capsid that modifies interactions with pattern recognition receptors and downstream signaling. In addition, the presence, generation, or secretion of extracellular proteins can indirectly influence the response to HAdVs via the activation and recruitment of cells at the site of infection.
Collapse
|
19
|
Soto JA, Gálvez NMS, Andrade CA, Pacheco GA, Bohmwald K, Berrios RV, Bueno SM, Kalergis AM. The Role of Dendritic Cells During Infections Caused by Highly Prevalent Viruses. Front Immunol 2020; 11:1513. [PMID: 32765522 PMCID: PMC7378533 DOI: 10.3389/fimmu.2020.01513] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are a type of innate immune cells with major relevance in the establishment of an adaptive response, as they are responsible for the activation of lymphocytes. Since their discovery, several reports of their role during infectious diseases have been performed, highlighting their functions and their mechanisms of action. DCs can be categorized into different subsets, and each of these subsets expresses a wide arrange of receptors and molecules that aid them in the clearance of invading pathogens. Interferon (IFN) is a cytokine -a molecule of protein origin- strongly associated with antiviral immune responses. This cytokine is secreted by different cell types and is fundamental in the modulation of both innate and adaptive immune responses against viral infections. Particularly, DCs are one of the most important immune cells that produce IFN, with type I IFNs (α and β) highlighting as the most important, as they are associated with viral clearance. Type I IFN secretion can be induced via different pathways, activated by various components of the virus, such as surface proteins or genetic material. These molecules can trigger the activation of the IFN pathway trough surface receptors, including IFNAR, TLR4, or some intracellular receptors, such as TLR7, TLR9, and TLR3. Here, we discuss various types of dendritic cells found in humans and mice; their contribution to the activation of the antiviral response triggered by the secretion of IFN, through different routes of the induction for this important antiviral cytokine; and as to how DCs are involved in human infections that are considered highly frequent nowadays.
Collapse
Affiliation(s)
- Jorge A Soto
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolas M S Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A Pacheco
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roslye V Berrios
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Baker AH, Herzog RW. Did Dendritic Cell Activation, Induced by Adenovirus-Antibody Complexes, Play a Role in the Death of Jesse Gelsinger? Mol Ther 2020; 28:704-706. [PMID: 32061269 DOI: 10.1016/j.ymthe.2020.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Andrew H Baker
- University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Roland W Herzog
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
21
|
Mennechet FJD, Paris O, Ouoba AR, Salazar Arenas S, Sirima SB, Takoudjou Dzomo GR, Diarra A, Traore IT, Kania D, Eichholz K, Weaver EA, Tuaillon E, Kremer EJ. A review of 65 years of human adenovirus seroprevalence. Expert Rev Vaccines 2019; 18:597-613. [PMID: 31132024 DOI: 10.1080/14760584.2019.1588113] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Human adenovirus (HAdV)-derived vectors have been used in numerous pre-clinical and clinical trials during the last 40 years. Current research in HAdV-based vaccines focuses on improving transgene immunogenicity and safety. Because pre-existing humoral immunity against HAdV types correlate with reduced vaccine efficacy and safety, many groups are exploring the development of HAdV types vectors with lower seroprevalence. However, global seroepidemiological data are incomplete. Areas covered: The goal of this review is to centralize 65 years of research on (primarily) HAdV epidemiology. After briefly addressing adenovirus biology, we chronical HAdV seroprevalence studies and highlight major milestones. Finally, we analyze data from about 50 studies with respect to HAdVs types that are currently used in the clinic, or are in the developmental pipeline. Expert opinion: Vaccination is among the most efficient tools to prevent infectious disease. HAdV-based vaccines have undeniable potential, but optimization is needed and antivector immunity remains a challenge if the same vectors are to be administrated to different populations. Here, we identify gaps in our knowledge and the need for updated worldwide epidemiological data.
Collapse
Affiliation(s)
- Franck J D Mennechet
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Océane Paris
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Aline Raissa Ouoba
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France.,b UMR 1058, Pathogenesis and Control of Chronic Infections , INSERM - University of Montpellier - Establishment Français du Sang - Centre Hospitalier Universitaire de Montpellier , Montpellier , France.,c Département des sciences et de la recherche clinique , Centre Muraz , Bobo-Dioulasso , Burkina Faso
| | - Sofia Salazar Arenas
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Sodiomon B Sirima
- d Centre National de Recherche et de Formation sur le Paludisme , Ouagadougou , Burkina Faso.,e Groupe de Recherche Action en Santé (GRAS) , Ouagadougou , Burkina Faso
| | - Guy R Takoudjou Dzomo
- f Complexe Hospitalo Universitaire « Le Bon Samaritain » , N'Djamena , Republic of Chad
| | - Amidou Diarra
- d Centre National de Recherche et de Formation sur le Paludisme , Ouagadougou , Burkina Faso
| | - Isidore T Traore
- c Département des sciences et de la recherche clinique , Centre Muraz , Bobo-Dioulasso , Burkina Faso
| | - Dramane Kania
- c Département des sciences et de la recherche clinique , Centre Muraz , Bobo-Dioulasso , Burkina Faso
| | - Karsten Eichholz
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| | - Eric A Weaver
- g University of Nebraska-Lincoln, School of Biological Sciences , Lincoln , NE , USA
| | - Edouard Tuaillon
- b UMR 1058, Pathogenesis and Control of Chronic Infections , INSERM - University of Montpellier - Establishment Français du Sang - Centre Hospitalier Universitaire de Montpellier , Montpellier , France
| | - Eric J Kremer
- a Institut de Génétique Moléculaire de Montpellier , University of Montpellier - CNRS , Montpellier , France
| |
Collapse
|
22
|
Agrawal B, Gupta N, Vedi S, Singh S, Li W, Garg S, Li J, Kumar R. Heterologous Immunity between Adenoviruses and Hepatitis C Virus (HCV): Recombinant Adenovirus Vaccine Vectors Containing Antigens from Unrelated Pathogens Induce Cross-Reactive Immunity Against HCV Antigens. Cells 2019; 8:E507. [PMID: 31130710 PMCID: PMC6562520 DOI: 10.3390/cells8050507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/13/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
Host immune responses play an important role in the outcome of infection with hepatitis C virus (HCV). They can lead to viral clearance and a positive outcome, or progression and severity of chronic disease. Extensive research in the past >25 years into understanding the immune responses against HCV have still resulted in many unanswered questions implicating a role for unknown factors and events. In our earlier studies, we made a surprising discovery that peptides derived from structural and non-structural proteins of HCV have substantial amino acid sequence homologies with various proteins of adenoviruses and that immunizing mice with a non-replicating, non-recombinant adenovirus vector leads to induction of a robust cross-reactive cellular and humoral response against various HCV antigens. In this work, we further demonstrate antibody cross-reactivity between Ad and HCV in vivo. We also extend this observation to show that recombinant adenoviruses containing antigens from unrelated pathogens also possess the ability to induce cross-reactive immune responses against HCV antigens along with the induction of transgene antigen-specific immunity. This cross-reactive immunity can (a) accommodate the making of dual-pathogen vaccines, (b) play an important role in the natural course of HCV infection and (c) provide a plausible answer to many unexplained questions regarding immunity to HCV.
Collapse
Affiliation(s)
- Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Nancy Gupta
- Department of Laboratory Medicine & Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Satish Vedi
- Department of Laboratory Medicine & Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Shakti Singh
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Wen Li
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Saurabh Garg
- Department of Laboratory Medicine & Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Jie Li
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| | - Rakesh Kumar
- Department of Laboratory Medicine & Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2S2, Canada.
| |
Collapse
|
23
|
del Rio D, Beucher B, Lavigne M, Wehbi A, Gonzalez Dopeso-Reyes I, Saggio I, Kremer EJ. CAV-2 Vector Development and Gene Transfer in the Central and Peripheral Nervous Systems. Front Mol Neurosci 2019; 12:71. [PMID: 30983967 PMCID: PMC6449469 DOI: 10.3389/fnmol.2019.00071] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/07/2019] [Indexed: 12/11/2022] Open
Abstract
The options available for genetic modification of cells of the central nervous system (CNS) have greatly increased in the last decade. The current panoply of viral and nonviral vectors provides multifunctional platforms to deliver expression cassettes to many structures and nuclei. These cassettes can replace defective genes, modify a given pathway perturbed by diseases, or express proteins that can be selectively activated by drugs or light to extinguish or excite neurons. This review focuses on the use of canine adenovirus type 2 (CAV-2) vectors for gene transfer to neurons in the brain, spinal cord, and peripheral nervous system. We discuss (1) recent advances in vector production, (2) why CAV-2 vectors preferentially transduce neurons, (3) the mechanism underlying their widespread distribution via retrograde axonal transport, (4) how CAV-2 vectors have been used to address structure/function, and (5) their therapeutic applications.
Collapse
Affiliation(s)
- Danila del Rio
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Bertrand Beucher
- PVM, BioCampus, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Marina Lavigne
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Amani Wehbi
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | | | - Isabella Saggio
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Institute of Structural Biology, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|