1
|
Lu HH, Dos Santos Alves RP, Li QH, Eder L, Timis J, Madany H, Chuensirikulchai K, Varghese KV, Singh A, Le Tran L, Street A, Elong Ngono A, Croft M, Shresta S. Enhanced durability of a Zika virus self-amplifying RNA vaccine through combinatorial OX40 and 4-1BB agonism. JCI Insight 2025; 10:e187405. [PMID: 40178907 DOI: 10.1172/jci.insight.187405] [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: 09/30/2024] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
The SARS-CoV-2 pandemic highlighted the potential of mRNA vaccines in rapidly responding to emerging pathogens. However, immunity induced by conventional mRNA vaccines wanes quickly, requiring frequent boosters. Self-amplifying RNA (saRNA) vaccines, which extend antigen expression via self-replication, offer a promising strategy to induce more durable immune responses. In this study, we developed an saRNA vaccine encoding Zika virus (ZIKV) membrane and envelope proteins and evaluated its efficacy in mice. A single vaccination elicited strong humoral and cellular immune responses and reduced viral loads but only for 28 days. By day 84, antibody titers and T cell responses had significantly declined, resulting in reduced efficacy. To address this, we evaluated agonist antibodies targeting the T cell costimulatory molecules OX40 and 4-1BB. Coadministration of agonist antibodies enhanced CD8+ T cell responses to vaccination, resulting in sustained immunity and reduced viral loads at day 84. Depletion and passive transfer studies verified that long-term antiviral immunity was primarily CD8+ T cell dependent, with minimal contributions from antibody responses. These findings suggest that agonists targeting members of the tumor necrosis receptor superfamily, such as OX40 and 4-1BB, might enhance the durability of saRNA vaccine-induced protection, addressing a key limitation of current mRNA vaccine platforms.
Collapse
Affiliation(s)
- Hsueh-Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
| | | | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
| | - Luke Eder
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Henry Madany
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Krithik V Varghese
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Aditi Singh
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Linda Le Tran
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Audrey Street
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Wang YT, Branche E, Xie J, McMillan RE, Ana-Sosa-Batiz F, Lu HH, Li QH, Clark AE, Valls Cuevas JM, Viramontes KM, Garretson AF, Dos Santos Alves RP, Heinz S, Benner C, Carlin AF, Shresta S. Zika but not Dengue virus infection limits NF-κB activity in human monocyte-derived dendritic cells and suppresses their ability to activate T cells. Nat Commun 2025; 16:2695. [PMID: 40133263 PMCID: PMC11937581 DOI: 10.1038/s41467-025-57977-2] [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: 08/17/2024] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Understanding flavivirus immunity is critical for the development of pan-flavivirus vaccines. Dendritic cells (DC) coordinate antiviral innate and adaptive immune responses, and they can be targeted by flaviviruses as a mechanism of immune evasion. Using an unbiased genome-wide approach designed to specifically identify flavivirus-modulated pathways, we found that, while dengue virus (DENV) robustly activates DCs, Zika virus (ZIKV) causes minimal activation of genes involved in DC activation, maturation, and antigen presentation, reducing cytokine secretion and the stimulation of allogeneic and peptide-specific T cell responses. Mechanistically, ZIKV inhibits DC maturation by suppressing NF-κB p65 recruitment and the subsequent transcription of proinflammatory and DC maturation-related genes. Thus, we identify a divergence in the effects of ZIKV and DENV on the host T cell response, highlighting the need to factor such differences into the design of anti-flavivirus vaccines.
Collapse
Affiliation(s)
- Ying-Ting Wang
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Emilie Branche
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jialei Xie
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Rachel E McMillan
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, La Jolla, California, USA
| | | | - Hsueh-Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, La Jolla, California, USA
| | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, La Jolla, California, USA
| | - Alex E Clark
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Joan M Valls Cuevas
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Karla M Viramontes
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Aaron F Garretson
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Sven Heinz
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Christopher Benner
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Aaron F Carlin
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Roth C, Pitard B, Levillayer L, Lay S, Vo HTM, Cantaert T, Sakuntabhai A. Zika virus T-cell based 704/DNA vaccine promotes protection from Zika virus infection in the absence of neutralizing antibodies. PLoS Negl Trop Dis 2024; 18:e0012601. [PMID: 39418312 PMCID: PMC11521268 DOI: 10.1371/journal.pntd.0012601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/29/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are closely related flaviviruses co-circulating in the same endemic areas. Infection can raise cross-reactive antibodies that can be either protective or increase risk of severe disease, depending on the infection sequence, DENV serotype and elapsed time between infection. On the contrast, T cell-mediated immunity against DENV and ZIKV is considered protective. Therefore, we have developed a T cell vaccine enriched in immunodominant T cell epitopes derived from ZIKV and evaluated its immunogenicity and efficacy against ZIKV and DENV infection. Mice were vaccinated using DNA vaccine platform using the tetrafunctional amphiphilic block copolymer 704. We show that vaccination of 2 different HLA class I transgenic mice with the ZIKV non-structural (NS) poly-epitope elicits T cell response against numerous ZIKV epitopes. Moreover, vaccination induces a significant protection against ZIKV infection, in the absence of neutralizing or enhancing antibodies against ZIKV. However, vaccination does not induce a significant protection against DENV2. In contrast, immunization with a DENV1-NS poly-epitope induces a significant protection against both DENV1 and DENV2, in the absence of humoral immunity. Taken together, we have shown that T-cell based vaccination could protect against multiple flavivirus infections and could overcome the complexity of antibody-mediated enhancement.
Collapse
Affiliation(s)
- Claude Roth
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| | - Bruno Pitard
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in Immunotherapy, INCIT UMR1232/EMR6001, F-44000 Nantes, France
| | - Laurine Levillayer
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Oxford University Clinical Research Unit, Ho Chi Minh, Vietnam
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Anavaj Sakuntabhai
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| |
Collapse
|
4
|
Porier DL, Adam A, Kang L, Michalak P, Tupik J, Santos MA, Tanelus M, López K, Auguste DI, Lee C, Allen IC, Wang T, Auguste AJ. Humoral and T-cell-mediated responses to an insect-specific flavivirus-based Zika virus vaccine candidate. PLoS Pathog 2024; 20:e1012566. [PMID: 39388457 PMCID: PMC11495591 DOI: 10.1371/journal.ppat.1012566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 10/22/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Flaviviruses represent a significant global health threat and relatively few licensed vaccines exist to protect against them. Insect-specific flaviviruses (ISFVs) are incapable of replication in humans and have emerged as a novel and promising tool for flavivirus vaccine development. ISFV-based flavivirus vaccines have shown exceptional safety, immunogenicity, and efficacy, however, a detailed assessment of the correlates of protection and immune responses induced by these vaccines are still needed for vaccine optimization. Here, we explore the mechanisms of protective immunity induced by a previously created pre-clinical Zika virus (ZIKV) vaccine candidate, called Aripo/Zika (ARPV/ZIKV). In brief, immunocompromised IFN-αβR-/- mice passively immunized with ARPV/ZIKV immune sera experienced protection after lethal ZIKV challenge, although this protection was incomplete. ARPV/ZIKV-vaccinated IFN-αβR-/- mice depleted of CD4+ or CD8+ T-cells at the time of ZIKV challenge showed no morbidity or mortality. However, the adoptive transfer of ARPV/ZIKV-primed T-cells into recipient IFN-αβR-/- mice resulted in a two-day median increase in survival time compared to controls. Altogether, these results suggest that ARPV/ZIKV-induced protection is primarily mediated by neutralizing antibodies at the time of challenge and that T-cells may play a comparatively minor but cumulative role in the protection observed. Lastly, ARPV/ZIKV-vaccinated Tcra KO mice, which are deficient in T-cell responses, experienced significant mortality post-challenge. These results suggest that ARPV/ZIKV-induced cell-mediated responses are critical for development of protective immune responses at vaccination. Despite the strong focus on neutralizing antibody responses to novel flavivirus vaccine candidates, these results suggest that cell-mediated responses induced by ISFV-based vaccines remain important to overall protective responses.
Collapse
Affiliation(s)
- Danielle L. Porier
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lin Kang
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, United States of America
| | - Pawel Michalak
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Juselyn Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
| | - Matthew A. Santos
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Manette Tanelus
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Krisangel López
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Dawn I. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Christy Lee
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Albert J. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|
5
|
Porier DL, Adam A, Kang L, Michalak P, Tupik J, Santos MA, Lee C, Allen IC, Wang T, Auguste AJ. Humoral and T-cell-mediated responses to a pre-clinical Zika vaccine candidate that utilizes a unique insect-specific flavivirus platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530296. [PMID: 36909623 PMCID: PMC10002724 DOI: 10.1101/2023.03.01.530296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Vaccination is critical for the control and prevention of viral outbreaks, yet conventional vaccine platforms may involve trade-offs between immunogenicity and safety. Insect-specific viruses have emerged as a novel vaccine platform to overcome this challenge. Detailed studies of humoral and T-cell responses induced by new insect-specific flavivirus (ISFV)-based vaccine platforms are needed to better understand correlates of protection and improve vaccine efficacy. Previously, we used a novel ISFV called Aripo virus (ARPV) to create a Zika virus (ZIKV) vaccine candidate (designated ARPV/ZIKV). ARPV/ZIKV demonstrated exceptional safety and single-dose efficacy, completely protecting mice from a lethal ZIKV challenge. Here, we explore the development of immune responses induced by ARPV/ZIKV immunization and evaluate its correlates of protection. Passive transfer of ARPV/ZIKV-induced immune sera to naïve mice prior to challenge emphasized the importance of neutralizing antibodies as a correlate of protection. Depletion of T-cells in vaccinated mice and adoptive transfer of ARPV/ZIKV-primed T-cells to naïve mice prior to challenge indicated that ARPV/ZIKV-induced CD4 + and CD8 + T-cell responses contribute to the observed protection but may not be essential for protection during ZIKV challenge. However, vaccination of Rag1 KO, Tcra KO, and muMt - mice demonstrated the critical role for ARPV/ZIKV-induced T-cells in developing protective immune responses following vaccination. Overall, both humoral and T-cell-mediated responses induced by ISFV-based vaccines are important for comprehensive immunity, and ISFV platforms continue to be a promising method for future vaccine development.
Collapse
|
6
|
Garcia G, Chakravarty N, Abu AE, Jeyachandran AV, Takano KA, Brown R, Morizono K, Arumugaswami V. Replication-Deficient Zika Vector-Based Vaccine Provides Maternal and Fetal Protection in Mouse Model. Microbiol Spectr 2022; 10:e0113722. [PMID: 36169338 PMCID: PMC9602260 DOI: 10.1128/spectrum.01137-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/28/2022] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne human pathogen, causes dire congenital brain developmental abnormalities in children of infected mothers. The global health crisis precipitated by this virus has led to a concerted effort to develop effective therapies and prophylactic measures although, unfortunately, not very successfully. The error-prone nature of RNA viral genome replication tends to promote evolution of novel viral strains, which could cause epidemics and pandemics. As such, our objective was to develop a safe and effective replication-deficient ZIKV vector-based vaccine candidate. We approached this by generating a ZIKV vector containing only the nonstructural (NS) 5'-untranslated (UTR)-NS-3' UTR sequences, with the structural proteins capsid (C), precursor membrane (prM), and envelope (E) (CprME) used as a packaging system. We efficiently packaged replication-deficient Zika vaccine particles in human producer cells and verified antigen expression in vitro. In vivo studies showed that, after inoculation in neonatal mice, the Zika vaccine candidate (ZVAX) was safe and did not produce any replication-competent revertant viruses. Immunization of adult, nonpregnant mice showed that ZVAX protected mice from lethal challenge by limiting viral replication. We then evaluated the safety and efficacy of ZVAX in pregnant mice, where it was shown to provide efficient maternal and fetal protection against Zika disease. Mass cytometry analysis showed that vaccinated pregnant animals had high levels of splenic CD8+ T cells and effector memory T cell responses with reduced proinflammatory cell responses, suggesting that endogenous expression of NS proteins by ZVAX induced cellular immunity against ZIKV NS proteins. We also investigated humoral immunity against ZIKV, which is potentially induced by viral proteins present in ZVAX virions. We found no significant difference in neutralizing antibody titer in vaccinated or unvaccinated challenged animals; therefore, it is likely that cellular immunity plays a major role in ZVAX-mediated protection against ZIKV infection. In conclusion, we demonstrated ZVAX as an effective inducer of protective immunity against ZIKV, which can be further evaluated for potential prophylactic application in humans. IMPORTANCE This research is important as it strives to address the critical need for effective prophylactic measures against the outbreak of Zika virus (ZIKV) and outlines an important vaccine technology that could potentially be used to induce immune responses against other pandemic-potential viruses.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Nikhil Chakravarty
- Department of Epidemiology, University of California, Los Angeles, Los Angeles, California, USA
| | - Angel Elma Abu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Kari-Ann Takano
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Rebecca Brown
- Departments of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Kouki Morizono
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
7
|
Borchmann S, Selenz C, Lohmann M, Ludwig H, Gassa A, Brägelmann J, Lohneis P, Meder L, Mattlener J, Breid S, Nill M, Fassunke J, Wisdom AJ, Compes A, Gathof B, Alakus H, Kirsch D, Hekmat K, Büttner R, Reinhardt HC, Hallek M, Ullrich RT. Tripartite antigen-agnostic combination immunotherapy cures established poorly immunogenic tumors. J Immunother Cancer 2022; 10:e004781. [PMID: 36223955 PMCID: PMC9562723 DOI: 10.1136/jitc-2022-004781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Single-agent immunotherapy has shown remarkable efficacy in selected cancer entities and individual patients. However, most patients fail to respond. This is likely due to diverse immunosuppressive mechanisms acting in a concerted way to suppress the host anti-tumor immune response. Combination immunotherapy approaches that are effective in such poorly immunogenic tumors mostly rely on precise knowledge of antigenic determinants on tumor cells. Creating an antigen-agnostic combination immunotherapy that is effective in poorly immunogenic tumors for which an antigenic determinant is not known is a major challenge. METHODS We use multiple cell line and poorly immunogenic syngeneic, autochthonous, and autologous mouse models to evaluate the efficacy of a novel combination immunotherapy named tripartite immunotherapy (TRI-IT). To elucidate TRI-ITs mechanism of action we use immune cell depletions and comprehensive tumor and immune infiltrate characterization by flow cytometry, RNA sequencing and diverse functional assays. RESULTS We show that combined adoptive cellular therapy (ACT) with lymphokine-activated killer cells, cytokine-induced killer cells, Vγ9Vδ2-T-cells (γδ-T-cells) and T-cells enriched for tumor recognition (CTLs) display synergistic antitumor effects, which are further enhanced by cotreatment with anti-PD1 antibodies. Most strikingly, the full TRI-IT protocol, a combination of this ACT with anti-PD1 antibodies, local immunotherapy of agonists against toll-like receptor 3, 7 and 9 and pre-ACT lymphodepletion, eradicates and induces durable anti-tumor immunity in a variety of poorly immunogenic syngeneic, autochthonous, as well as autologous humanized patient-derived models. Mechanistically, we show that TRI-IT coactivates adaptive cellular and humoral, as well as innate antitumor immune responses to mediate its antitumor effect without inducing off-target toxicity. CONCLUSIONS Overall, TRI-IT is a novel, highly effective, antigen-agnostic, non-toxic combination immunotherapy. In this study, comprehensive insights into its preclinical efficacy, even in poorly immunogenic tumors, and mode of action are given, so that translation into clinical trials is the next step.
Collapse
Affiliation(s)
- Sven Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Carolin Selenz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Mia Lohmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Hanna Ludwig
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Asmae Gassa
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Johannes Brägelmann
- Mildred Scheel School of Oncology, University Hospital Cologne, Medical Faculty, Cologne, Germany
| | - Philipp Lohneis
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Lydia Meder
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Julia Mattlener
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Sara Breid
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Marieke Nill
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Amy J Wisdom
- Department of Radiation Oncology and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Anik Compes
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Birgit Gathof
- Institute of Transfusion Medicine, University of Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - David Kirsch
- Department of Radiation Oncology and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Khosro Hekmat
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | | | - H Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen,University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Roland T Ullrich
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
8
|
ZIKV-envelope proteins induce specific humoral and cellular immunity in distinct mice strains. Sci Rep 2022; 12:15733. [PMID: 36131132 PMCID: PMC9492693 DOI: 10.1038/s41598-022-20183-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Recent outbreaks of Zika virus (ZIKV) infection have highlighted the need for a better understanding of ZIKV-specific immune responses. The ZIKV envelope glycoprotein (EZIKV) is the most abundant protein on the virus surface and it is the main target of the protective immune response. EZIKV protein contains the central domain (EDI), a dimerization domain containing the fusion peptide (EDII), and a domain that binds to the cell surface receptor (EDIII). In this study, we performed a systematic comparison of the specific immune response induced by different EZIKV recombinant proteins (EZIKV, EDI/IIZIKV or EDIIIZIKV) in two mice strains. Immunization induced high titers of E-specific antibodies which recognized ZIKV-infected cells and neutralized the virus. Furthermore, immunization with EZIKV, EDI/IIZIKV and EDIIIZIKV proteins induced specific IFNγ-producing cells and polyfunctional CD4+ and CD8+ T cells. Finally, we identified 4 peptides present in the envelope protein (E1-20, E51-70, E351-370 and E361-380), capable of inducing a cellular immune response to the H-2Kd and H-2Kb haplotypes. In summary, our work provides a detailed assessment of the immune responses induced after immunization with different regions of the ZIKV envelope protein.
Collapse
|
9
|
Marzan-Rivera N, Serrano-Collazo C, Cruz L, Pantoja P, Ortiz-Rosa A, Arana T, Martinez MI, Burgos AG, Roman C, Mendez LB, Geerling E, Pinto AK, Brien JD, Sariol CA. Infection order outweighs the role of CD4 + T cells in tertiary flavivirus exposure. iScience 2022; 25:104764. [PMID: 35982798 PMCID: PMC9379573 DOI: 10.1016/j.isci.2022.104764] [Citation(s) in RCA: 3] [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: 04/18/2022] [Revised: 06/12/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022] Open
Abstract
The link between CD4+ T and B cells during immune responses to DENV and ZIKV and their roles in cross-protection during heterologous infection is an active area of research. Here we used CD4+ lymphocyte depletions to dissect the impact of cellular immunity on humoral responses during a tertiary flavivirus infection in macaques. We show that CD4+ depletion in DENV/ZIKV-primed animals followed by DENV resulted in dysregulated adaptive immune responses. We show a delay in DENV-specific IgM/IgG antibody titers and binding and neutralization in the DENV/ZIKV-primed CD4-depleted animals but not in ZIKV/DENV-primed CD4-depleted animals. This study confirms the critical role of CD4+ cells in priming an early effective humoral response during sequential flavivirus infections. Our work here suggests that the order of flavivirus exposure affects the outcome of a tertiary infection. Our findings have implications for understanding the complex flavivirus immune responses and for the development of effective flavivirus vaccines.
Collapse
Affiliation(s)
- Nicole Marzan-Rivera
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Crisanta Serrano-Collazo
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Lorna Cruz
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Petraleigh Pantoja
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Alexandra Ortiz-Rosa
- Department of Biology, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA
| | - Teresa Arana
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Melween I. Martinez
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Caribbean Primate Research Center, School of Medicine, University of Puerto Rico-Medical Sciences Campus, Toa Baja, PR 00952, USA
| | - Armando G. Burgos
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Caribbean Primate Research Center, School of Medicine, University of Puerto Rico-Medical Sciences Campus, Toa Baja, PR 00952, USA
| | - Chiara Roman
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Loyda B. Mendez
- Department of Science & Technology, Universidad Ana G. Mendez, Recinto de Carolina, Carolina, PR 00985, USA
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 631204, USA
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 631204, USA
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 631204, USA
| | - Carlos A. Sariol
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Department of Biology, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA
- Department of Internal Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| |
Collapse
|
10
|
Zhou Y, Bian P, Du H, Wang T, Li M, Hu H, Ye C, Zheng X, Zhang Y, Lei Y, Jia Z, Lian J. The Comparison of Inflammatory Cytokines (IL-6 and IL-18) and Immune Cells in Japanese Encephalitis Patients With Different Progression. Front Cell Infect Microbiol 2022; 12:826603. [PMID: 35463639 PMCID: PMC9022626 DOI: 10.3389/fcimb.2022.826603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Background Japanese encephalitis virus (JEV) is the main cause of viral encephalitis in Asia. Nowadays, no effective and specific therapy for JE patients is available except supportive treatment. The fatality rate of JE patients is still about 30%, and more than half of survivors suffered from various neuropsychiatric sequelae. Thus, more attention should be paid to JE. Methods In this study, a retrospective cohort of JE patients was collected and the general features of JE patients admitted into the Department of Infectious Diseases were analyzed. Meanwhile, the dynamic change of plasma cytokines and immune cells in JE patients with divergent prognosis was detected and analyzed. Results We found a mounted proportion of adult/old patients in JE cases. The level of IL-6 and IL-18 increased in JE patients especially in fatal individuals. There was a continuous decreased percentage of CD4+ T and B cells in severe JE patients with fatal outcome compared with the surviving JE patients. Conclusions The consistent high level of IL-6 and IL-18 in the plasma and low proportion of CD4+ T and B cells in the PBMCs might be the indicators of poor prognosis.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Peiyu Bian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Hong Du
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Tao Wang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Mengyuan Li
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haifeng Hu
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xuyang Zheng
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Yinfeng Lei
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi’an, China
| | - Zhansheng Jia
- The Center of Infectious Diseases and Liver, Xi’an International Medical Center Hospital, Xi’an, China
- *Correspondence: Zhansheng Jia, ; Jianqi Lian,
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Zhansheng Jia, ; Jianqi Lian,
| |
Collapse
|
11
|
Pardy RD, Gentile ME, Carter AM, Condotta SA, King IL, Richer MJ. An Epidemic Zika Virus Isolate Drives Enhanced T Follicular Helper Cell and B Cell-Mediated Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1719-1728. [PMID: 35346966 PMCID: PMC8976755 DOI: 10.4049/jimmunol.2100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 01/27/2022] [Indexed: 11/19/2022]
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen that recently caused a series of increasingly severe outbreaks. We previously demonstrated that, compared with a pre-epidemic isolate (ZIKVCDN), a Brazilian ZIKV isolate (ZIKVBR) possesses a novel capacity to suppress host immunity, resulting in delayed viral clearance. However, whether ZIKVBR modulates CD4 T cell responses remains unknown. In this study, we show that, in comparison with ZIKVCDN infection, CD4 T cells are less polarized to the Th1 subtype following ZIKVBR challenge in mice. In contrast, we observed an enhanced accumulation of T follicular helper cells 10, 14, and 21 d postinfection with ZIKVBR This response correlated with an enhanced germinal center B cell response and robust production of higher avidity-neutralizing Abs following ZIKVBR infection. Taken together, our data suggest that contemporary ZIKV strains have evolved to differentially induce CD4 T cell, B cell, and Ab responses and this could provide a model to further define the signals required for T follicular helper cell development.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Maria E Gentile
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University Health Centre, McGill University, Montreal, Quebec, Canada; and
| | - Alexandria M Carter
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Stephanie A Condotta
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Irah L King
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University Health Centre, McGill University, Montreal, Quebec, Canada; and
| | - Martin J Richer
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada;
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
12
|
Santiago HC, Pereira-Neto TA, Gonçalves-Pereira MH, Terzian ACB, Durbin AP. Peculiarities of Zika Immunity and Vaccine Development: Lessons from Dengue and the Contribution from Controlled Human Infection Model. Pathogens 2022; 11:pathogens11030294. [PMID: 35335618 PMCID: PMC8951202 DOI: 10.3390/pathogens11030294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 01/27/2023] Open
Abstract
The Zika virus (ZIKV) was first isolated from a rhesus macaque in the Zika forest of Uganda in 1947. Isolated cases were reported until 2007, when the first major outbreaks of Zika infection were reported from the Island of Yap in Micronesia and from French Polynesia in 2013. In 2015, ZIKV started to circulate in Latin America, and in 2016, ZIKV was considered by WHO to be a Public Health Emergency of International Concern due to cases of Congenital Zika Syndrome (CZS), a ZIKV-associated complication never observed before. After a peak of cases in 2016, the infection incidence dropped dramatically but still causes concern because of the associated microcephaly cases, especially in regions where the dengue virus (DENV) is endemic and co-circulates with ZIKV. A vaccine could be an important tool to mitigate CZS in endemic countries. However, the immunological relationship between ZIKV and other flaviviruses, especially DENV, and the low numbers of ZIKV infections are potential challenges for developing and testing a vaccine against ZIKV. Here, we discuss ZIKV vaccine development with the perspective of the immunological concerns implicated by DENV-ZIKV cross-reactivity and the use of a controlled human infection model (CHIM) as a tool to accelerate vaccine development.
Collapse
Affiliation(s)
- Helton C. Santiago
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 30270-901, MG, Brazil; (T.A.P.-N.); (M.H.G.-P.)
- Correspondence: ; Tel.: +55-31-3409-2664
| | - Tertuliano A. Pereira-Neto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 30270-901, MG, Brazil; (T.A.P.-N.); (M.H.G.-P.)
| | - Marcela H. Gonçalves-Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 30270-901, MG, Brazil; (T.A.P.-N.); (M.H.G.-P.)
| | - Ana C. B. Terzian
- Laboratory of Cellular Immunology, Rene Rachou Institute, Fiocruz, Belo Horizonte 30190-002, MG, Brazil;
| | - Anna P. Durbin
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| |
Collapse
|
13
|
Evaluation of the Levels of Peripheral CD3 +, CD4 +, and CD8 + T Cells and IgG and IgM Antibodies in COVID-19 Patients at Different Stages of Infection. Microbiol Spectr 2022; 10:e0084521. [PMID: 35196808 PMCID: PMC8865559 DOI: 10.1128/spectrum.00845-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection affects the stimulatory levels of cellular-mediated immunity, which plays an essential role in controlling SARS-CoV-2 infection. In fact, several studies have shown the association of lymphopenia with severe COVID-19 in patients. The aim of this study is to investigate the response of the immune system, including cell-mediated immunity and antibody production, during different stages of SARS-CoV-2 infection. Peripheral blood and serum samples were collected from patients with moderate infection, patients under medication (hospitalized), patients who had recovered, and healthy individuals (n = 80). Flow cytometry analysis was performed on peripheral blood samples to determine the cellular immunity profile of each patient. The data showed a significant reduction in the levels of CD3+, CD4+, and CD8+ T cells and CD45+ cells in the moderate and under-medication groups, suggesting lymphopenia in those patients. Also, enzyme-linked immunosorbent assay (ELISA) was conducted on the serum samples to measure the levels of antibodies, including IgM and IgG, in each patient. The results revealed a significant increase in the levels of IgM in the moderate infection and under-medication patients, thus indicating the production of IgM during the first week of infection. Furthermore, changes in the levels of IgG were significantly detected among recovered patients, indicating therefore a remarkable increase during the recovery stage of SARS-CoV-2 infection and thus a strong humoral-mediated immunity. In summary, the results of this study may help us to understand the main role of the cellular immune responses, including CD3+, CD4+, and CD8+ T cells, against SARS-CoV-2 infection. This understanding might support the development of SARS-CoV-2 treatments and vaccines in the near future. IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late 2019 in China. This virus is a serious threat to people not only in China but also worldwide, where it has been detected in over 222 countries. It has been reported that ∼3.4% of SARS-CoV-2-infected patients have died. The significance of our study relies on the fact that an enzyme-linked immunosorbent assay and flow cytometry were used to measure the levels of antibodies and cellular immune response, respectively, from clinical samples of patients infected with SARS-CoV-2.
Collapse
|
14
|
Stone ET, Hassert M, Geerling E, Wagner C, Brien JD, Ebel GD, Hirsch AJ, German C, Smith JL, Pinto AK. Balanced T and B cell responses are required for immune protection against Powassan virus in virus-like particle vaccination. Cell Rep 2022; 38:110388. [PMID: 35172138 PMCID: PMC8919300 DOI: 10.1016/j.celrep.2022.110388] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/07/2021] [Accepted: 01/24/2022] [Indexed: 12/29/2022] Open
Abstract
Powassan virus (POWV) is a tick-borne pathogen for which humans are an incidental host. POWV infection can be fatal or result in long-term neurological sequelae; however, there are no approved vaccinations for POWV. Integral to efficacious vaccine development is the identification of correlates of protection, which we accomplished in this study by utilizing a murine model of POWV infection. Using POWV lethal and sub-lethal challenge models, we show that (1) robust B and T cell responses are necessary for immune protection, (2) POWV lethality can be attributed to both viral- and host-mediated drivers of disease, and (3) knowledge of the immune correlates of protection against POWV can be applied in a virus-like particle (VLP)-based vaccination approach that provides protection from lethal POWV challenge. Identification of these immune protection factors is significant as it will aid in the rational design of POWV vaccines.
Collapse
Affiliation(s)
- E Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - Colleen Wagner
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - Gregory D Ebel
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Alec J Hirsch
- The Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA; Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Cody German
- The Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA; Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jessica L Smith
- The Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA; Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63104, USA.
| |
Collapse
|
15
|
Lee HN, Manangeeswaran M, Lewkowicz AP, Engel K, Chowdhury M, Garige M, Eckhaus MA, Sourbier C, Ireland DD, Verthelyi D. NK cells require immune checkpoint receptor LILRB4/gp49B to control neurotropic Zika virus infections in mice. JCI Insight 2022; 7:151420. [PMID: 35132958 PMCID: PMC8855830 DOI: 10.1172/jci.insight.151420] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Immune cells express an array of inhibitory checkpoint receptors that are upregulated upon activation and limit tissue damage associated with excessive response to pathogens or allergens. Mouse leukocyte immunoglobulin like receptor B4 (LILRB4), also known as glycoprotein 49B (gp49B), is an inhibitory checkpoint receptor constitutively expressed in myeloid cells and upregulated in B cells, T cells, and NK cells upon activation. Here, we report that expression of LILRB4, which binds Zika virus (ZIKV), was increased in microglia and myeloid cells infiltrating the brains of neonatal mice with ZIKV-associated meningoencephalitis. Importantly, while C57BL/6 mice developed transient neurological symptoms but survived infection, mice lacking LILRB4/gp49B (LILRB4 KO) exhibited more severe signs of neurological disease and succumbed to disease. Their brains showed increased cellular infiltration but reduced control of viral burden. The reduced viral clearance was associated with altered NK cell function in the absence of LILRB4/gp49B. In naive animals, this manifested as reduced granzyme B responses to stimulation, but in ZIKV-infected animals, NK cells showed phenotypic changes that suggested altered maturation, diminished glucose consumption, reduced IFN-γ and granzyme B production, and impaired cytotoxicity. Together, our data reveal LILRB4/gp49B as an important regulator of NK cell function during viral infections.
Collapse
Affiliation(s)
- Ha-Na Lee
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Mohanraj Manangeeswaran
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Aaron P Lewkowicz
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Kaliroi Engel
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Monica Chowdhury
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Mamatha Garige
- Laboratory of Molecular Oncology, Division of Biotechnology Review and Research-I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Michael A Eckhaus
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Carole Sourbier
- Laboratory of Molecular Oncology, Division of Biotechnology Review and Research-I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Derek Dc Ireland
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Daniela Verthelyi
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| |
Collapse
|
16
|
Mangare C, Tischer-Zimmermann S, Bonifacius A, Riese SB, Dragon AC, Blasczyk R, Maecker-Kolhoff B, Eiz-Vesper B. Variances in Antiviral Memory T-Cell Repertoire of CD45RA- and CD62L-Depleted Lymphocyte Products Reflect the Need of Individual T-Cell Selection Strategies to Reduce the Risk of GvHD while Preserving Antiviral Immunity in Adoptive T-Cell Therapy. Transfus Med Hemother 2022; 49:30-43. [PMID: 35221866 PMCID: PMC8832244 DOI: 10.1159/000516284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/01/2021] [Indexed: 04/03/2025] Open
Abstract
INTRODUCTION Viral infections and reactivations still remain a cause of morbidity and mortality after hematopoietic stem cell transplantation due to immunodeficiency and immunosuppression. Transfer of unmanipulated donor-derived lymphocytes (DLI) represents a promising strategy for improving cellular immunity but carries the risk of graft versus host disease (GvHD). Depleting alloreactive naïve T cells (TN) from DLIs was implemented to reduce the risk of GvHD induction while preserving antiviral memory T-cell activity. Here, we compared two TN depletion strategies via CD45RA and CD62L expression and investigated the presence of antiviral memory T cells against human adenovirus (AdV) and Epstein-Barr virus (EBV) in the depleted fractions in relation to their functional and immunophenotypic characteristics. METHODS T-cell responses against ppEBV_EBNA1, ppEBV_Consensus and ppAdV_Hexon within TN-depleted (CD45RA-/CD62L-) and TN-enriched (CD45RA+/CD62L+) fractions were quantified by interferon-gamma (IFN-γ) ELISpot assay after short- and long-term in vitro stimulation. T-cell frequencies and immunophenotypic composition were assessed in all fractions by flow cytometry. Moreover, alloimmune T-cell responses were evaluated by mixed lymphocyte reaction. RESULTS According to differences in the phenotype composition, antigen-specific T-cell responses in CD45RA- fraction were up to 2 times higher than those in the CD62L- fraction, with the highest increase (up to 4-fold) observed after 7 days for ppEBV_EBNA1-specific T cells. The CD4+ effector memory T cells (TEM) were mainly responsible for EBV_EBNA1- and AdV_Hexon-specific T-cell responses, whereas the main functionally active T cells against ppEBV_Consensus were CD8+ central memory T cells (TCM) and TEM. Moreover, comparison of both depletion strategies indicated that alloreactivity in CD45RA- was lower than that in CD62L- fraction. CONCLUSION Taken together, our results indicate that CD45RA depletion is a more suitable strategy for generating TN-depleted products consisting of memory T cells against ppEBV_EBNA1 and ppAdV_Hexon than CD62L in terms of depletion effectiveness, T-cell functionality and alloreactivity. To maximally exploit the beneficial effects mediated by antiviral memory T cells in TN-depleted products, depletion methods should be selected individually according to phenotype composition and CD4/CD8 antigen restriction. TN-depleted DLIs may improve the clinical outcome in terms of infections, GvHD, and disease relapse if selection of pathogen-specific donor T cells is not available.
Collapse
Affiliation(s)
- Caroline Mangare
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sebastian B. Riese
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Anna Christina Dragon
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Shin M, Kim K, Lee HJ, Lee R, Jung YJ, Park J, Hahn TW. Zika virus baculovirus-expressed envelope protein elicited humoral and cellular immunity in immunocompetent mice. Sci Rep 2022; 12:660. [PMID: 35027643 PMCID: PMC8758750 DOI: 10.1038/s41598-021-04713-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/24/2021] [Indexed: 12/05/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that has a high risk of inducing Guillain-Barré syndrome and microcephaly in newborns. Because vaccination is considered the most effective strategy against ZIKV infection, we designed a recombinant vaccine utilizing the baculovirus expression system with two strains of ZIKV envelope protein (MR766, Env_M; ZBRX6, Env_Z). Animals inoculated with Env_M and Env_Z produced ZIKV-specific antibodies and secreted effector cytokines such as interferon-γ, tumor necrosis factor-α, and interleukin-12. Moreover, the progeny of immunized females had detectable maternal antibodies that protected them against two ZIKV strains (MR766 and PRVABC59) and a Dengue virus strain. We propose that the baculovirus expression system ZIKV envelope protein recombinant provides a safe and effective vaccine strategy.
Collapse
Affiliation(s)
- Minna Shin
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Kiju Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyo-Ji Lee
- College of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rangyeon Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Yu-Jin Jung
- College of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jeongho Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Tae-Wook Hahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
18
|
Bhatia B, Meade-White K, Haddock E, Feldmann F, Marzi A, Feldmann H. A live-attenuated viral vector vaccine protects mice against lethal challenge with Kyasanur Forest disease virus. NPJ Vaccines 2021; 6:152. [PMID: 34907224 PMCID: PMC8671490 DOI: 10.1038/s41541-021-00416-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/18/2021] [Indexed: 12/02/2022] Open
Abstract
Kyasanur Forest disease virus (KFDV) is a tick-borne flavivirus endemic in India known to cause severe hemorrhagic and encephalitic disease in humans. In recent years, KFDV has spread beyond its original endemic zone raising public health concerns. Currently, there is no treatment available for KFDV but a vaccine with limited efficacy is used in India. Here, we generated two new KFDV vaccine candidates based on the vesicular stomatitis virus (VSV) platform. We chose the VSV-Ebola virus (VSV-EBOV) vector either with the full-length or a truncated EBOV glycoprotein as the vehicle to express the precursor membrane (prM) and envelope (E) proteins of KFDV (VSV-KFDV). For efficacy testing, we established a mouse disease model by comparing KFDV infections in three immunocompetent mouse strains (BALB/c, C57Bl/6, and CD1). Both vaccine vectors provided promising protection against lethal KFDV challenge in the BALB/c model following prime-only prime-boost and immunizations. Only prime-boost immunization with VSV-KFDV expressing full-length EBOV GP resulted in uniform protection. Hyperimmune serum derived from prime-boost immunized mice protected naïve BALB/c mice from lethal KFDV challenge indicating the importance of antibodies for protection. The new VSV-KFDV vectors are promising vaccine candidates to combat an emerging, neglected public health problem in a densely populated part of the world.
Collapse
Affiliation(s)
- Bharti Bhatia
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
19
|
Cavazzoni CB, Bozza VB, Lucas TC, Conde L, Maia B, Mesin L, Schiepers A, Ersching J, Neris RL, Conde JN, Coelho DR, Lima TM, Alvim RG, Castilho LR, de Paula Neto HA, Mohana-Borges R, Assunção-Miranda I, Nobrega A, Victora GD, Vale AM. The immunodominant antibody response to Zika virus NS1 protein is characterized by cross-reactivity to self. J Exp Med 2021; 218:e20210580. [PMID: 34292314 PMCID: PMC8302445 DOI: 10.1084/jem.20210580] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Besides antigen-specific responses to viral antigens, humoral immune response in virus infection can generate polyreactive and autoreactive antibodies. Dengue and Zika virus infections have been linked to antibody-mediated autoimmune disorders, including Guillain-Barré syndrome. A unique feature of flaviviruses is the secretion of nonstructural protein 1 (NS1) by infected cells. NS1 is highly immunogenic, and antibodies targeting NS1 can have both protective and pathogenic roles. In the present study, we investigated the humoral immune response to Zika virus NS1 and found NS1 to be an immunodominant viral antigen associated with the presence of autoreactive antibodies. Through single B cell cultures, we coupled binding assays and BCR sequencing, confirming the immunodominance of NS1. We demonstrate the presence of self-reactive clones in germinal centers after both infection and immunization, some of which present cross-reactivity with NS1. Sequence analysis of anti-NS1 B cell clones showed sequence features associated with pathogenic autoreactive antibodies. Our findings demonstrate NS1 immunodominance at the cellular level as well as a potential role for NS1 in ZIKV-associated autoimmune manifestations.
Collapse
Affiliation(s)
- Cecilia B. Cavazzoni
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Vicente B.T. Bozza
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tostes C.V. Lucas
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Maia
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Jonatan Ersching
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Romulo L.S. Neris
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas N. Conde
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego R. Coelho
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tulio M. Lima
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata G.F. Alvim
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leda R. Castilho
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. de Paula Neto
- Laboratório de Alvos Moleculares, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Nobrega
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Andre M. Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Sun J, Zheng Z, Li M, Liu Z, Su X, Jin X. Development of a novel ZIKV vaccine comprised of immunodominant CD4+ and CD8+ T cell epitopes identified through comprehensive epitope mapping in Zika virus infected mice. Vaccine 2021; 39:5173-5186. [PMID: 34353682 DOI: 10.1016/j.vaccine.2021.07.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 11/28/2022]
Abstract
Zika virus (ZIKV) caused over two million human infections in more than 80 countries around 2015-2016. Current vaccines under development are mostly focused on inducing antibodies that despite capable of inhibiting the virus, may have the potential to trigger antibody dependent enhancement (ADE). T cell vaccines that do not induce antibodies targeting viral surface will unlikely cause ADE, but be capable of potentiating the effectiveness of an antibody-inducing vaccine. To develop such a protective T cell vaccine, we first examined the repertoire of antigen-specific T cells in immunocompetent mice that have been transiently infected by ZIKV. Through epitope mapping using 427 overlapping peptides spanning the entire length of ZIKV polyprotein, we discovered 27 immunodominant epitopes scattered throughout the virus on C, E, NS1-NS5 proteins. Among them, 8 were confirmed as CD4+ T cell epitopes, and 16 as CD8+ T cell epitopes, while 3 for both T cell subsets. From these 27 newly identified epitopes, the top 10 epitopes were selected to formulate three T cell vaccines comprised of either CD4+ T cell epitopes, or CD8+ T cell epitopes, or a mixture of both. Immunization with these T cell epitopes induced T cell-mediated cytotoxicity and cytokine production, and conferred varying degrees of protection against ZIKV challenge. Moreover, these new T cell vaccines also improved the protective efficacy of a neutralizing antibody-inducing recombinant E80 protein vaccine. Together, our results provided additional evidence in support of the protective role of ZIKV-specific CD4+ and CD8+ T cells, and laid foundation for future development of T cell vaccines for ZIKV.
Collapse
Affiliation(s)
- Jin Sun
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhihang Zheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Min Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Zhihua Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Xiao Su
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xia Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China.
| |
Collapse
|
21
|
da Silva Antunes R, Pallikkuth S, Williams E, Dawen Yu E, Mateus J, Quiambao L, Wang E, Rawlings SA, Stadlbauer D, Jiang K, Amanat F, Arnold D, Andrews D, Fuego I, Dan JM, Grifoni A, Weiskopf D, Krammer F, Crotty S, Hoffer ME, Pahwa SG, Sette A. Differential T-Cell Reactivity to Endemic Coronaviruses and SARS-CoV-2 in Community and Health Care Workers. J Infect Dis 2021; 224:70-80. [PMID: 33822097 PMCID: PMC8083569 DOI: 10.1093/infdis/jiab176] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Herein we measured CD4+ T-cell responses against common cold coronaviruses (CCC) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in high-risk health care workers (HCW) and community controls. We observed higher levels of CCC-reactive T cells in SARS-CoV-2-seronegative HCW compared to community donors, consistent with potential higher occupational exposure of HCW to CCC. We further show that SARS-CoV-2 T-cell reactivity of seronegative HCW was higher than community controls and correlation between CCC and SARS-CoV-2 responses is consistent with cross-reactivity and not associated with recent in vivo activation. Surprisingly, CCC T-cell reactivity was decreased in SARS-CoV-2-infected HCW, suggesting that exposure to SARS-CoV-2 might interfere with CCC responses, either directly or indirectly. This result was unexpected, but consistently detected in independent cohorts derived from Miami and San Diego. CD4+ T-cell responses against common cold coronaviruses (CCC) are elevated in SARS-CoV-2 seronegative high-risk health care workers (HCW) compared to COVID-19 convalescent HCW, suggesting that exposure to SARS-CoV-2 might interfere with CCC responses and/or cross-reactivity associated with a protective effect.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Erin Williams
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Lorenzo Quiambao
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Stephen A Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David Arnold
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David Andrews
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Irma Fuego
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| | - Michael E Hoffer
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Savita G Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
22
|
CD4 + T Cells Cross-Reactive with Dengue and Zika Viruses Protect against Zika Virus Infection. Cell Rep 2021; 31:107566. [PMID: 32348763 PMCID: PMC7261136 DOI: 10.1016/j.celrep.2020.107566] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 02/06/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022] Open
Abstract
The underlying mechanisms by which prior immunity to dengue virus (DENV) affords cross-protection against the related flavivirus Zika virus (ZIKV) are poorly understood. Here, we examine the ability of DENV/ZIKV-cross-reactive CD4+ T cells to protect against versus exacerbate ZIKV infection by using a histocompatibility leukocyte antigen (HLA)-DRB1*0101 transgenic, interferon α/β receptor-deficient mouse model that supports robust DENV and ZIKV replication. By mapping the HLA-DRB1*0101-restricted T cell response, we identify DENV/ZIKV-cross-reactive CD4+ T cell epitopes that stimulate interferon gamma (IFNγ) and/or tumor necrosis factor (TNF) production. Vaccination of naive HLA-DRB1*0101 transgenic mice with these peptides induces a CD4+ T cell response sufficient to reduce tissue viral burden following ZIKV infection. Notably, this protective response requires IFNγ and/or TNF secretion but not anti-ZIKV immunoglobulin G (IgG) production. Thus, DENV/ZIKV-cross-reactive CD4+ T cells producing canonical Th1 cytokines can suppress ZIKV replication in an antibody-independent manner. These results may have important implications for increasing the efficacy and safety of DENV/ZIKV vaccines and for developing pan-flavivirus vaccines.
Collapse
|
23
|
Repeated exposure to dengue virus elicits robust cross neutralizing antibodies against Zika virus in residents of Northeastern Thailand. Sci Rep 2021; 11:9634. [PMID: 33953258 PMCID: PMC8100282 DOI: 10.1038/s41598-021-88933-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/19/2021] [Indexed: 11/23/2022] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are antigenically related mosquito-borne flaviviruses. ZIKV is becoming increasingly prevalent in DENV-endemic regions, raising the possibility that pre-existing immunity to one virus could modulate the response to a heterologous virus, although whether this would be beneficial or detrimental is unclear. Here, we analyzed sera from residents of a DENV-endemic region of Thailand to determine the prevalence of DENV-elicited antibodies capable of cross-neutralizing ZIKV. Sixty-one participants who were asymptomatic and unselected for viral serostatus were enrolled. Among them, 52 and 51 were seropositive for IgG antibody against DENV or ZIKV E proteins (ELISA assay), respectively. Notably, 44.23% (23/52) of DENV seropositive participants had serological evidence of multiple exposures to DENV, and these subjects had strikingly higher titers and broader reactivities of neutralizing antibodies (NAbs) against ZIKV and DENV heterotypes compared with participants with serological evidence of a single DENV infection (25/52, 48.1%). In total, 17 of the 61 participants (27.9%) had NAbs against ZIKV and all four DENV serotypes, and an additional 9 (14.8%) had NAbs against ZIKV and DENV1, 2, and 3. NAbs against DENV2 were the most prevalent (44/61, 72.1%) followed by DENV3 (38/61, 62.3%) and DENV1 (36/61, 59.0%). Of note, anti-ZIKV NAbs were more prevalent than anti-DENV4 NAbs (27/61, 44.3% and 21/61, 34.4%, respectively). Primary ZIKV infection was detected in two participants, confirming that ZIKV co-circulates in this region. Thus, residents of DENV-endemic regions with repeated exposure to DENV have higher titers of NAbs against ZIKV than individuals with only a single DENV exposure.
Collapse
|
24
|
Zika Virus Pathogenesis: A Battle for Immune Evasion. Vaccines (Basel) 2021; 9:vaccines9030294. [PMID: 33810028 PMCID: PMC8005041 DOI: 10.3390/vaccines9030294] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infection and its associated congenital and other neurological disorders, particularly microcephaly and other fetal developmental abnormalities, constitute a World Health Organization (WHO) Zika Virus Research Agenda within the WHO’s R&D Blueprint for Action to Prevent Epidemics, and continue to be a Public Health Emergency of International Concern (PHEIC) today. ZIKV pathogenicity is initiated by viral infection and propagation across multiple placental and fetal tissue barriers, and is critically strengthened by subverting host immunity. ZIKV immune evasion involves viral non-structural proteins, genomic and non-coding RNA and microRNA (miRNA) to modulate interferon (IFN) signaling and production, interfering with intracellular signal pathways and autophagy, and promoting cellular environment changes together with secretion of cellular components to escape innate and adaptive immunity and further infect privileged immune organs/tissues such as the placenta and eyes. This review includes a description of recent advances in the understanding of the mechanisms underlying ZIKV immune modulation and evasion that strongly condition viral pathogenesis, which would certainly contribute to the development of anti-ZIKV strategies, drugs, and vaccines.
Collapse
|
25
|
Immunity to TBEV Related Flaviviruses with Reduced Pathogenicity Protects Mice from Disease but Not from TBEV Entry into the CNS. Vaccines (Basel) 2021; 9:vaccines9030196. [PMID: 33652698 PMCID: PMC7996866 DOI: 10.3390/vaccines9030196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is a leading cause of vector-borne viral encephalitis with expanding endemic regions across Europe. In this study we tested in mice the efficacy of preinfection with a closely related low-virulent flavivirus, Langat virus (LGTV strain TP21), or a naturally avirulent TBEV strain (TBEV-280) in providing protection against lethal infection with the highly virulent TBEV strain (referred to as TBEV-Hypr). We show that prior infection with TP21 or TBEV-280 is efficient in protecting mice from lethal TBEV-Hypr challenge. Histopathological analysis of brains from nonimmunized mice revealed neuronal TBEV infection and necrosis. Neuroinflammation, gliosis, and neuronal necrosis was however also observed in some of the TP21 and TBEV-280 preinfected mice although at reduced frequency as compared to the nonimmunized TBEV-Hypr infected mice. qPCR detected the presence of viral RNA in the CNS of both TP21 and TBEV-280 immunized mice after TBEV-Hypr challenge, but significantly reduced compared to mock-immunized mice. Our results indicate that although TBEV-Hypr infection is effectively controlled in the periphery upon immunization with low-virulent LGTV or naturally avirulent TBEV 280, it may still enter the CNS of these animals. These findings contribute to our understanding of causes for vaccine failure in individuals vaccinated with TBE vaccines.
Collapse
|
26
|
Lima NS, Moon D, Darko S, De La Barrera RA, Lin L, Koren MA, Jarman RG, Eckels KH, Thomas SJ, Michael NL, Modjarrad K, Douek DC, Trautmann L. Pre-existing Immunity to Japanese Encephalitis Virus Alters CD4 T Cell Responses to Zika Virus Inactivated Vaccine. Front Immunol 2021; 12:640190. [PMID: 33717194 PMCID: PMC7943459 DOI: 10.3389/fimmu.2021.640190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
The epidemic spread of Zika virus (ZIKV), associated with devastating neurologic syndromes, has driven the development of multiple ZIKV vaccines candidates. An effective vaccine should induce ZIKV-specific T cell responses, which are shown to improve the establishment of humoral immunity and contribute to viral clearance. Here we investigated how previous immunization against Japanese encephalitis virus (JEV) and yellow fever virus (YFV) influences T cell responses elicited by a Zika purified-inactivated virus (ZPIV) vaccine. We demonstrate that three doses of ZPIV vaccine elicited robust CD4 T cell responses to ZIKV structural proteins, while ZIKV-specific CD4 T cells in pre-immunized individuals with JEV vaccine, but not YFV vaccine, were more durable and directed predominantly toward conserved epitopes, which elicited Th1 and Th2 cytokine production. In addition, T cell receptor repertoire analysis revealed preferential expansion of cross-reactive clonotypes between JEV and ZIKV, suggesting that pre-existing immunity against JEV may prime the establishment of stronger CD4 T cell responses to ZPIV vaccination. These CD4 T cell responses correlated with titers of ZIKV-neutralizing antibodies in the JEV pre-vaccinated group, but not in flavivirus-naïve or YFV pre-vaccinated individuals, suggesting a stronger contribution of CD4 T cells in the generation of neutralizing antibodies in the context of JEV-ZIKV cross-reactivity.
Collapse
Affiliation(s)
- Noemia S Lima
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Cellular Immunology Section, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Damee Moon
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Samuel Darko
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rafael A De La Barrera
- Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Leyi Lin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael A Koren
- Viral Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Richard G Jarman
- Viral Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Kenneth H Eckels
- Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Stephen J Thomas
- Division of Infectious Diseases, Department of Medicine, State University of New York Upstate, Syracuse, NY, United States
| | - Nelson L Michael
- Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lydie Trautmann
- Cellular Immunology Section, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
27
|
da Silva Antunes R, Pallikkuth S, Williams E, Yu ED, Mateus J, Quiambao L, Wang E, Rawlings SA, Stadlbauer D, Jiang K, Amanat F, Arnold D, Andrews D, Fuego I, Dan JM, Grifoni A, Weiskopf D, Krammer F, Crotty S, Hoffer ME, Pahwa SG, Sette A. Differential T cell reactivity to seasonal coronaviruses and SARS-CoV-2 in community and health care workers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.12.21249683. [PMID: 33469594 PMCID: PMC7814840 DOI: 10.1101/2021.01.12.21249683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Herein we measured CD4+ T cell responses against common cold corona (CCC) viruses and SARS-CoV-2 in high-risk health care workers (HCW) and community controls. We observed higher levels of CCC reactive T cells in SARS-CoV-2 seronegative HCW compared to community donors, consistent with potential higher occupational exposure of HCW to CCC. We further show that SARS-CoV-2 reactivity of seronegative HCW was higher than community controls and correlation between CCC and SARS-CoV-2 responses is consistent with cross-reactivity and not associated with recent in vivo activation. Surprisingly, CCC reactivity was decreased in SARS-CoV-2 infected HCW, suggesting that exposure to SARS-CoV-2 might interfere with CCC responses, either directly or indirectly. This result was unexpected, but consistently detected in independent cohorts derived from Miami and San Diego.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Erin Williams
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Lorenzo Quiambao
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Stephen A. Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Arnold
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - David Andrews
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Irma Fuego
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Jennifer M. Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Michael E. Hoffer
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Savita G. Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
28
|
Baron MD, Hodgson S, Moffat K, Qureshi M, Graham SP, Darpel KE. Depletion of CD8 + T cells from vaccinated goats does not affect protection from challenge with wild-type peste des petits ruminants virus. Transbound Emerg Dis 2020; 68:3320-3334. [PMID: 33222411 PMCID: PMC9291567 DOI: 10.1111/tbed.13936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022]
Abstract
Peste des petits ruminants (PPR) is a severe disease of goats and sheep that is widespread in Africa, the Middle East and Asia. The disease is caused by peste des petits ruminants virus (PPRV); cell culture-attenuated strains of PPRV have been shown, both experimentally and by extensive use in the field, to be effective vaccines and are widely used. We have previously demonstrated that these vaccines elicit both serological (PPRV-specific antibody) and cell-based (PPRV-specific CD4+ and CD8+ T cells) immune responses. However, it is not known which of these responses are required for protection from PPRV, information that would be useful in the evaluation of new vaccines that are being developed to provide the capability to differentiate infected and vaccinated animals (DIVA capability). To begin to address this issue, we have used a complement-fixing monoclonal antibody recognizing caprine CD8 to deplete >99.9% of circulating CD8+ T cells from vaccinated goats. Animals were then infected with wild-type PPRV. Despite the absence of the CD8+ T-cell component of the vaccine-induced immune response, the vaccinated animals were almost fully protected, showing no pyrexia or viraemia, and almost no clinical signs. These data suggest that a virus-specific CD8+ T-cell response is not critical for protection against PPRV and that virus-specific antibody and/or CD4+ T cells are the main mediators of protection. We have also shown that the leucopenia caused by infection with wild-type PPRV affects all major classes of circulating leucocytes.
Collapse
Affiliation(s)
| | - Sophia Hodgson
- The Pirbright Institute, Pirbright, UK.,School of Veterinary Medicine, University of Surrey, Guildford, UK
| | | | | | - Simon P Graham
- The Pirbright Institute, Pirbright, UK.,School of Veterinary Medicine, University of Surrey, Guildford, UK
| | | |
Collapse
|
29
|
Elong Ngono A, Syed T, Nguyen AV, Regla-Nava JA, Susantono M, Spasova D, Aguilar A, West M, Sparks J, Gonzalez A, Branche E, DeHart JL, Vega JB, Karmali PP, Chivukula P, Kamrud K, Aliahmad P, Wang N, Shresta S. CD8 + T cells mediate protection against Zika virus induced by an NS3-based vaccine. SCIENCE ADVANCES 2020; 6:eabb2154. [PMID: 33148638 PMCID: PMC7673678 DOI: 10.1126/sciadv.abb2154] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/21/2020] [Indexed: 05/07/2023]
Abstract
Zika virus (ZIKV) is associated with congenital malformations in infants born to infected mothers, and with Guillain-Barré syndrome in infected adults. Development of ZIKV vaccines has focused predominantly on the induction of neutralizing antibodies, although a suboptimal antibody response may theoretically enhance disease severity through antibody-dependent enhancement (ADE). Here, we report induction of a protective anti-ZIKV CD8+ T cell response in the HLA-B*0702 Ifnar1-/- transgenic mice using an alphavirus-based replicon RNA vaccine expressing ZIKV nonstructural protein NS3, a potent T cell antigen. The NS3 vaccine did not induce a neutralizing antibody response but elicited polyfunctional CD8+ T cells that were necessary and sufficient for preventing death in lethally infected adult mice and fetal growth restriction in infected pregnant mice. These data identify CD8+ T cells as the major mediators of ZIKV NS3 vaccine-induced protection and suggest a new strategy to develop safe and effective anti-flavivirus vaccines.
Collapse
Affiliation(s)
- Annie Elong Ngono
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Thasneem Syed
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Anh-Viet Nguyen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Jose Angel Regla-Nava
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Mercylia Susantono
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Darina Spasova
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Allison Aguilar
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Melissa West
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Jessica Sparks
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Andrew Gonzalez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Emilie Branche
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Jason L DeHart
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Jerel Boyd Vega
- Arcturus Therapeutics Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Priya Prakash Karmali
- Arcturus Therapeutics Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Padmanabh Chivukula
- Arcturus Therapeutics Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Kurt Kamrud
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Parinaz Aliahmad
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Nathaniel Wang
- Synthetic Genomics Inc., 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sujan Shresta
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| |
Collapse
|
30
|
Stephenson KE, Tan CS, Walsh SR, Hale A, Ansel JL, Kanjilal DG, Jaegle K, Peter L, Borducchi EN, Nkolola JP, Makoni T, Fogel R, Bradshaw C, Tyler A, Moseley E, Chandrashekar A, Yanosick KE, Seaman MS, Eckels KH, De La Barrera RA, Thompson J, Dawson P, Thomas SJ, Michael NL, Modjarrad K, Barouch DH. Safety and immunogenicity of a Zika purified inactivated virus vaccine given via standard, accelerated, or shortened schedules: a single-centre, double-blind, sequential-group, randomised, placebo-controlled, phase 1 trial. THE LANCET. INFECTIOUS DISEASES 2020; 20:1061-1070. [PMID: 32618279 PMCID: PMC7472641 DOI: 10.1016/s1473-3099(20)30085-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/23/2019] [Accepted: 02/07/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND The development of an effective vaccine against Zika virus remains a public health priority. A Zika purified inactivated virus (ZPIV) vaccine candidate has been shown to protect animals against Zika virus challenge and to be well tolerated and immunogenic in humans up to 8 weeks of follow-up. We aimed to assess the safety and immunogenicity of ZPIV in humans up to 52 weeks of follow-up when given via standard or accelerated vaccination schedules. METHODS We did a single-centre, double-blind, randomised controlled, phase 1 trial in healthy adults aged 18-50 years with no known history of flavivirus vaccination or infection at Beth Israel Deaconess Medical Center in Boston, MA, USA. Participants were sequentially enrolled into one of three groups: ZPIV given at weeks 0 and 4 (standard regimen), weeks 0 and 2 (accelerated regimen), or week 0 alone (single-dose regimen). Within each group, participants were randomly assigned using a computer-generated randomisation schedule to receive an intramuscular injection of 5 μg ZPIV or saline placebo, in a ratio of 5:1. The sponsor, clinical staff, investigators, participants, and laboratory personnel were masked to treatment assignment. The primary endpoint was safety up to day 364 after final dose administration, and secondary endpoints were proportion of participants with positive humoral immune responses (50% microneutralisation titre [MN50] ≥100) and geometric mean MN50 at observed peak response (ie, the highest neutralising antibody level observed for an individual participant across all timepoints) and week 28. All participants who received at least one dose of ZPIV or placebo were included in the safety population; the analysis of immunogenicity at observed peak included all participants who received at least one dose of ZPIV or placebo and had any adverse events or immunogenicity data after dosing. The week 28 immunogenicity analysis population consisted of all participants who received ZPIV or placebo and had immunogenicity data available at week 28. This trial is registered with ClinicalTrials.gov, NCT02937233. FINDINGS Between Dec 8, 2016, and May 17, 2017, 12 participants were enrolled into each group and then randomly assigned to vaccine (n=10) or placebo (n=2). There were no serious or grade 3 treatment-related adverse events. The most common reactions among the 30 participants who received the vaccine were injection-site pain (24 [80%]), fatigue (16 [53%]), and headache (14 [46%]). A positive response at observed peak titre was detected in all participants who received ZPIV via the standard regimen, in eight (80%) of ten participants who received ZPIV via the accelerated regimen, and in none of the ten participants who received ZPIV via the single-dose regimen. The geometric mean of all individual participants' observed peak values was 1153·9 (95% CI 455·2-2925·2) in the standard regimen group, 517·7 (142·9-1875·6) in the accelerated regimen group, and 6·3 (3·7-10·8) in the single-dose regimen group. At week 28, a positive response was observed in one (13%) of eight participants who received ZPIV via the standard regimen and in no participant who received ZPIV via the accelerated (n=7) or single-dose (n=10) regimens. The geomteric mean titre (GMT) at this timepoint was 13·9 (95% CI 3·5-55·1) in the standard regimen group and 6·9 (4·0-11·9) in the accelerated regimen group; antibody titres were undetectable at 28 weeks in participants who received ZPIV via the single-dose regimen. For all vaccine schedules, GMTs peaked 2 weeks after the final vaccination and declined to less than 100 by study week 16. There was no difference in observed peak GMTs between the standard 4-week and the accelerated 2-week boosting regimens (p=0·4494). INTERPRETATION ZPIV was safe and well tolerated in humans up to 52 weeks of follow-up. ZPIV immunogenicity required two doses and was not durable. Additional studies of ZPIV to optimise dosing schedules are ongoing. FUNDING The Henry M Jackson Foundation for the Advancement of Military Medicine.
Collapse
Affiliation(s)
- Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Chen Sabrina Tan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Stephen R Walsh
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Andrew Hale
- University of Vermont Medical Center, Burlington, VT, USA; Larner College of Medicine, Burlington, VT, USA
| | - Jessica L Ansel
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Diane G Kanjilal
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kate Jaegle
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Lauren Peter
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Joseph P Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tatenda Makoni
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rachel Fogel
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Connor Bradshaw
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Anna Tyler
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Edward Moseley
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Katherine E Yanosick
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
31
|
Kubinski M, Beicht J, Gerlach T, Volz A, Sutter G, Rimmelzwaan GF. Tick-Borne Encephalitis Virus: A Quest for Better Vaccines against a Virus on the Rise. Vaccines (Basel) 2020; 8:E451. [PMID: 32806696 PMCID: PMC7564546 DOI: 10.3390/vaccines8030451] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV), a member of the family Flaviviridae, is one of the most important tick-transmitted viruses in Europe and Asia. Being a neurotropic virus, TBEV causes infection of the central nervous system, leading to various (permanent) neurological disorders summarized as tick-borne encephalitis (TBE). The incidence of TBE cases has increased due to the expansion of TBEV and its vectors. Since antiviral treatment is lacking, vaccination against TBEV is the most important protective measure. However, vaccination coverage is relatively low and immunogenicity of the currently available vaccines is limited, which may account for the vaccine failures that are observed. Understanding the TBEV-specific correlates of protection is of pivotal importance for developing novel and improved TBEV vaccines. For affording robust protection against infection and development of TBE, vaccines should induce both humoral and cellular immunity. In this review, the adaptive immunity induced upon TBEV infection and vaccination as well as novel approaches to produce improved TBEV vaccines are discussed.
Collapse
Affiliation(s)
- Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Jana Beicht
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany;
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University (LMU) Munich, Veterinaerstr. 13, 80539 Munich, Germany;
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| |
Collapse
|
32
|
Campbell VL, Nguyen L, Snoey E, McClurkan CL, Laing KJ, Dong L, Sette A, Lindestam Arlehamn CS, Altmann DM, Boyton RJ, Roby JA, Gale M, Stone M, Busch MP, Norris PJ, Koelle DM. Proteome-Wide Zika Virus CD4 T Cell Epitope and HLA Restriction Determination. Immunohorizons 2020; 4:444-453. [PMID: 32753403 PMCID: PMC7839664 DOI: 10.4049/immunohorizons.2000068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 02/04/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen that caused an epidemic in 2015-2016. ZIKV-specific T cell responses are functional in animal infection models, and helper CD4 T cells promote avid Abs in the vaccine context. The small volumes of blood available from field research limit the determination of T cell epitopes for complex microbes such as ZIKV. The goal of this project was efficient determination of human ZIKV CD4 T cell epitopes at the whole proteome scale, including validation of reactivity to whole pathogen, using small blood samples from convalescent time points when T cell response magnitude may have waned. Polyclonal enrichment of candidate ZIKV-specific CD4 T cells used cell-associated virus, documenting that T cells in downstream peptide analyses also recognize whole virus after Ag processing. Sequential query of bulk ZIKV-reactive CD4 T cells with pooled/single ZIKV peptides and molecularly defined APC allowed precision epitope and HLA restriction assignments across the ZIKV proteome and enabled discovery of numerous novel ZIKV CD4 T cell epitopes. The research workflow is useful for the study of emerging infectious diseases with a very limited human blood sample availability.
Collapse
Affiliation(s)
| | - LeAnn Nguyen
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Elise Snoey
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Kerry J. Laing
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lichun Dong
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA,Department of Medicine, University of California-San Diego, La Jolla, CA, USA
| | | | - Danny M. Altmann
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Rosemary J. Boyton
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Justin A. Roby
- Center for Innate Immunity of Immune Disease, Department of Immunology, University of Washington, Seattle, WA, USA
| | - Michael Gale
- Center for Innate Immunity of Immune Disease, Department of Immunology, University of Washington, Seattle, WA, USA,Department of Global Health, University of Washington, Seattle, WA, USA,Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA,Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, California, USA,Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Phillip J. Norris
- Vitalant Research Institute, San Francisco, California, USA,Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - David M. Koelle
- Department of Medicine, University of Washington, Seattle, WA, USA,Department of Global Health, University of Washington, Seattle, WA, USA,Benaroya Research Institute, Seattle, WA, USA,Department of Laboratory Medicine, Seattle, WA, USA,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Corresponding author: David Koelle MD, 750 Republican Street, Room E651, Seattle, WA, 981109, phone 206 616 1940, fax 206 616 4898,
| |
Collapse
|
33
|
Pereira Neto TA, Gonçalves-Pereira MH, de Queiroz CP, Ramos MF, de Oliveira FDFS, Oliveira-Prado R, do Nascimento VA, Abdalla LF, Santos JHA, Martins-Filho OA, Naveca FG, Teixeira-Carvalho A, Santiago HDC. Multifunctional T cell response in convalescent patients two years after ZIKV infection. J Leukoc Biol 2020; 108:1265-1277. [PMID: 32726884 DOI: 10.1002/jlb.4ma0520-708r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Zika is an important emerging infectious disease in which the role of T cells remains elusive. This study aimed to evaluate the phenotype of multifunctional T cells in individuals 2 yr after exposure to Zika virus (ZIKV). We used a library of 671 synthetic peptides covering the whole polyprotein of ZIKV in pools corresponding to each viral protein (i.e., capsid, membrane precursor or prM, envelope, NS1 [nonstructural protein], NS2A + NS2B, NS3, NS4A + NS4B, and NS5) to stimulate PBMCs from individuals previously exposed to ZIKV. We observed an increased frequency of ZIKV-specific IFNγ, IL-17A, TNF, and IL-10 production by T cell populations. IFNγ and TNF production were especially stimulated by prM, capsid, or NS1 in CD8+ T cells and by capsid or prM in CD4+ T cells. In addition, there was an increase in the frequency of IL-10+ CD8+ T cells after stimulation with prM, capsid, NS1, NS3, or NS5. Multifunctional properties were observed in ZIKV-specific T cells responding especially to prM, capsid, NS1 or, to a smaller extent, NS3 antigens. For example, we found a consistent IFNγ + TNF+ CD8+ T cell population in response to most virus antigens and CD4+ and CD8+ T cells that were IFNγ + IL-17A+ and IL-17A+IL-10+, which could also produce TNF, in response to capsid, prM, NS1, or NS3 stimulation. Interestingly, CD8+ T cells were more prone to a multifunctional phenotype than CD4+ T cells, and multifunctional T cells were more efficient at producing cytokines than single-function cells. This work provides relevant insights into the quality of ZIKV-specific T cell responses and ZIKV immunity.
Collapse
Affiliation(s)
| | | | - Camila Pereira de Queiroz
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michele Faria Ramos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | - Felipe Gomes Naveca
- Leonidas e Maria Deane Institute, Oswaldo Cruz Foundation, Manaus, Amazonas, Brazil
| | | | - Helton da Costa Santiago
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
34
|
Prow NA, Liu L, McCarthy MK, Walters K, Kalkeri R, Geiger J, Koide F, Cooper TH, Eldi P, Nakayama E, Diener KR, Howley PM, Hayball JD, Morrison TE, Suhrbier A. The vaccinia virus based Sementis Copenhagen Vector vaccine against Zika and chikungunya is immunogenic in non-human primates. NPJ Vaccines 2020; 5:44. [PMID: 32550013 PMCID: PMC7265471 DOI: 10.1038/s41541-020-0191-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/24/2020] [Indexed: 01/09/2023] Open
Abstract
The Sementis Copenhagen Vector (SCV) is a new vaccinia virus-derived, multiplication-defective, vaccine technology assessed herein in non-human primates. Indian rhesus macaques (Macaca mulatta) were vaccinated with a multi-pathogen recombinant SCV vaccine encoding the structural polyproteins of both Zika virus (ZIKV) and chikungunya virus (CHIKV). After one vaccination, neutralising antibody responses to ZIKV and four strains of CHIKV, representative of distinct viral genotypes, were generated. A second vaccination resulted in significant boosting of neutralising antibody responses to ZIKV and CHIKV. Following challenge with ZIKV, SCV-ZIKA/CHIK-vaccinated animals showed significant reductions in viremias compared with animals that had received a control SCV vaccine. Two SCV vaccinations also generated neutralising and IgG ELISA antibody responses to vaccinia virus. These results demonstrate effective induction of immunity in non-human primates by a recombinant SCV vaccine and illustrates the utility of SCV as a multi-disease vaccine platform capable of delivering multiple large immunogens.
Collapse
Affiliation(s)
- Natalie A Prow
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029 Australia.,Australian Infectious Disease Research Centre, Brisbane, QLD 4029 and 4072 Australia.,Experimental Therapeutics Laboratory, School of Pharmacy and Medical Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5000 Australia
| | - Liang Liu
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5000 Australia
| | - Mary K McCarthy
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Kevin Walters
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701 USA
| | - Raj Kalkeri
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701 USA
| | - Jillian Geiger
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701 USA
| | - Fusataka Koide
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701 USA
| | - Tamara H Cooper
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5000 Australia
| | - Preethi Eldi
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5000 Australia
| | - Eri Nakayama
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029 Australia.,Department of Virology I, National Institute of Infectious Diseases, Tokyo, 162-8640 Japan
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5000 Australia.,Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005 Australia
| | | | - John D Hayball
- Experimental Therapeutics Laboratory, School of Pharmacy and Medical Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5000 Australia
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Andreas Suhrbier
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029 Australia.,Australian Infectious Disease Research Centre, Brisbane, QLD 4029 and 4072 Australia
| |
Collapse
|
35
|
Vaccine development during global epidemics: the Zika experience. THE LANCET. INFECTIOUS DISEASES 2020; 20:998-999. [PMID: 32618278 PMCID: PMC7202844 DOI: 10.1016/s1473-3099(20)30360-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022]
|
36
|
Lee CYP, Carissimo G, Chen Z, Lum FM, Abu Bakar F, Rajarethinam R, Teo TH, Torres-Ruesta A, Renia L, Ng LF. Type I interferon shapes the quantity and quality of the anti-Zika virus antibody response. Clin Transl Immunology 2020; 9:e1126. [PMID: 32346479 PMCID: PMC7184064 DOI: 10.1002/cti2.1126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives Zika virus (ZIKV) is a mosquito-borne flavivirus that re-emerged in 2015. The association between ZIKV and neurological complications initiated the development of relevant animal models to understand the mechanisms underlying ZIKV-induced pathologies. Transient inhibition of the type I interferon (IFN) pathway through the use of an IFNAR1-blocking antibody, MAR1-5A3, could efficiently permit active virus replication in immunocompetent animals. Type I IFN signalling is involved in the regulation of humoral responses, and thus, it is crucial to investigate the potential effects of type I IFN blockade towards B-cell responses. Methods In this study, comparative analysis was conducted using serum samples collected from ZIKV-infected wild-type (WT) animals either administered with or without MAR1-5A3. Results Serological assays revealed a more robust ZIKV-specific IgG response and subtype switching upon inhibition of type I IFN due to the abundance of antigen availability. This observation was corroborated by an increase in germinal centres, plasma cells and germinal centre B cells. Interestingly, although both groups of animals recognised different B-cell linear epitopes in the E and NS1 regions, there was no difference in neutralising capacity. Further characterisation of these epitopes in the E protein revealed a detrimental role of antibodies that were generated in the absence of type I IFN. Conclusion This study highlights the role of type I IFN in shaping the anti-ZIKV antibody response to generate beneficial antibodies and will help guide development of better vaccine candidates triggering efficient neutralising antibodies and avoiding detrimental ones.
Collapse
Affiliation(s)
- Cheryl Yi-Pin Lee
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,NUS Graduate School for Integrative Sciences and Engineering National University of Singapore Singapore
| | - Guillaume Carissimo
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore
| | - Zheyuan Chen
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,School of Medicine Dentistry & Biomedical Sciences Queen's University Belfast Belfast UK
| | - Fok-Moon Lum
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore
| | - Farhana Abu Bakar
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,School of Biological Sciences Nanyang Technological University Singapore Singapore
| | - Ravisankar Rajarethinam
- Institute of Molecular and Cell Biology Agency of Science, Technology and Research (ASTAR) Singapore
| | - Teck-Hui Teo
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,Present address: Institut Pasteur Unite de Pathogenie Microbienne Moleculaire Paris France
| | - Anthony Torres-Ruesta
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,Department of Biochemistry Yong Loo Lin School of Medicine National University of Singapore Singapore
| | - Laurent Renia
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore
| | - Lisa Fp Ng
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,Department of Biochemistry Yong Loo Lin School of Medicine National University of Singapore Singapore.,Institute of Infection and Global Health University of Liverpool Liverpool UK
| |
Collapse
|
37
|
Sun J, Du S, Zheng Z, Cheng G, Jin X. Defeat Dengue and Zika Viruses With a One-Two Punch of Vaccine and Vector Blockade. Front Microbiol 2020; 11:362. [PMID: 32265852 PMCID: PMC7100368 DOI: 10.3389/fmicb.2020.00362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 02/18/2020] [Indexed: 01/07/2023] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are two mosquito-borne flaviviruses afflicting nearly half of the world population. Human infection by these viruses can either be asymptomatic or manifest as clinical diseases from mild to severe. Despite more cases are presented as self-limiting febrile illness, severe dengue disease can be manifested as hemorrhagic fever and hemorrhagic shock syndrome, and ZIKV infection has been linked to increased incidence of peripheral neuropathy Guillain-Barre syndrome and central neural disease such as microcephaly. The current prevention and treatment of these infectious diseases are either non-satisfactory or entirely lacking. Because DENV and ZIKV have much similarities in genomic and structural features, almost identical mode of mosquito-mediated transmission, and probably the same pattern of host innate and adaptive immunity toward them, it is reasonable and often desirable to investigate these two viruses side-by-side, and thereby devise common countermeasures against both. Here, we review the existing knowledge on DENV and ZIKV regarding epidemiology, molecular virology, protective immunity and vaccine development, discuss recent new discoveries on the functions of flavivirus NS1 protein in viral pathogenesis and transmission, and propose a one-two punch strategy using vaccine and vector blockade to overcome antibody-dependent enhancement and defeat Dengue and Zika viruses.
Collapse
Affiliation(s)
- Jin Sun
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Senyan Du
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Zhihang Zheng
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China,Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xia Jin
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China,*Correspondence: Xia Jin, ;
| |
Collapse
|
38
|
An optimized purified inactivated Zika vaccine provides sustained immunogenicity and protection in cynomolgus macaques. NPJ Vaccines 2020; 5:19. [PMID: 32194996 PMCID: PMC7067768 DOI: 10.1038/s41541-020-0167-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/14/2020] [Indexed: 01/07/2023] Open
Abstract
The recent spread of Zika virus (ZIKV) through the Americas and Caribbean and its devastating consequences for pregnant women and their babies have driven the search for a safe and efficacious ZIKV vaccine. Among the vaccine candidates, a first-generation ZIKV purified inactivated vaccine (ZPIV), adjuvanted with aluminum hydroxide, developed by the Walter Reed Army Institute of Research (WRAIR), has elicited high seroconversion rates in participants in three phase-I clinical trials. In collaboration with the WRAIR, Sanofi Pasteur (SP) optimized the production scale, culture and purification conditions, and increased the regulatory compliance, both of which are critical for clinical development and licensure of this vaccine. Using a clinical batch of the first-generation ZPIV as a benchmark, we report that different doses of the optimized vaccine (ZPIV-SP) elicited sustained neutralizing antibodies, specific T- and memory B-cells, and provided complete protection against a ZIKV challenge in cynomolgus macaques. These data provide evidence that the ZPIV-SP vaccine performs at least as well as the ZPIV vaccine, and provide support for continued development in the event of future ZIKV outbreaks.
Collapse
|
39
|
Beneficial and Detrimental Effects of Regulatory T Cells in Neurotropic Virus Infections. Int J Mol Sci 2020; 21:ijms21051705. [PMID: 32131483 PMCID: PMC7084400 DOI: 10.3390/ijms21051705] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023] Open
Abstract
Neurotropic viruses infect the central nervous system (CNS) and cause acute or chronic neurologic disabilities. Regulatory T cells (Treg) play a critical role for immune homeostasis, but may inhibit pathogen-specific immunity in infectious disorders. The present review summarizes the current knowledge about Treg in human CNS infections and their animal models. Besides dampening pathogen-induced immunopathology, Treg have the ability to facilitate protective responses by supporting effector T cell trafficking to the infection site and the development of resident memory T cells. Moreover, Treg can reduce virus replication by inducing apoptosis of infected macrophages and attenuate neurotoxic astrogliosis and pro-inflammatory microglial responses. By contrast, detrimental effects of Treg are caused by suppression of antiviral immunity, allowing for virus persistence and latency. Opposing disease outcomes following Treg manipulation in different models might be attributed to differences in technique and timing of intervention, infection route, genetic background, and the host’s age. In addition, mouse models of virus-induced demyelination revealed that Treg are able to reduce autoimmunity and immune-mediated CNS damage in a disease phase-dependent manner. Understanding the unique properties of Treg and their complex interplay with effector cells represents a prerequisite for the development of new therapeutic approaches in neurotropic virus infections.
Collapse
|
40
|
Reynolds CJ, Watber P, Santos CNO, Ribeiro DR, Alves JC, Fonseca ABL, Bispo AJB, Porto RLS, Bokea K, de Jesus AMR, de Almeida RP, Boyton RJ, Altmann DM. Strong CD4 T Cell Responses to Zika Virus Antigens in a Cohort of Dengue Virus Immune Mothers of Congenital Zika Virus Syndrome Infants. Front Immunol 2020; 11:185. [PMID: 32132999 PMCID: PMC7040481 DOI: 10.3389/fimmu.2020.00185] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Background: There is an urgent need to understand the complex relationship between cross-reactive anti-viral immunity, disease susceptibility, and severity in the face of differential exposure to related, circulating Flaviviruses. Co-exposure to Dengue virus and Zika virus in Brazil is a case in point. A devastating aspect of the 2015-2016 South American Zika outbreak was the dramatic increase in numbers of infants born with microcephaly to mothers exposed to Zika virus during pregnancy. It has been proposed that this is more likely to ensue from Zika infection in women lacking cross-protective Dengue immunity. In this case series we measure the prevalence of Dengue immunity in a cohort of mothers exposed to Zika virus during pregnancy in the 2015-2016 Zika outbreak that gave birth to an infant affected by microcephaly and explore their adaptive immunity to Zika virus. Results: Fifty women from Sergipe, Brazil who gave birth to infants with microcephaly following Zika virus exposure during the 2015-16 outbreak were tested for serological evidence of Dengue exposure and IFNγ ELISpot spot forming cell (SFC) response to Zika virus. The majority (46/50) demonstrated Dengue immunity. IFNγ ELISpot responses to Zika virus antigens showed the following hierarchy: Env>NS1>NS3>C protein. Twenty T cell epitopes from Zika virus Env were identified. Responses to Zika virus antigens Env and NS1 were polyfunctional with cells making IFNγ, TNFα, IL-4, IL-13, and IL-10. In contrast, responses to NS5 only produced the immune regulatory TGFβ1 cytokine. There were SFC responses against Zika virus Env (1-20) and variant peptide sequences from West Nile virus, Dengue virus 1-4 and Yellow Fever virus. Conclusion: Almost all the women in our study showed serological evidence of Dengue immunity, suggesting that microcephaly can occur in DENV immune mothers. T cell immunity to Zika virus showed a multifunctional response to the antigens Env and NS1 and immune regulatory responses to NS5 and C protein. Our data support an argument that different viral products may skew the antiviral response to a more pro or anti-inflammatory outcome, with an associated impact on immunopathogenesis.
Collapse
Affiliation(s)
- Catherine J. Reynolds
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Patricia Watber
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Camilla Natália Oliveira Santos
- Molecular Biology Laboratory, Graduate Program in Health Science, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Danielle Rodrigues Ribeiro
- Molecular Biology Laboratory, Graduate Program in Health Science, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Juliana Cardoso Alves
- Molecular Biology Laboratory, Graduate Program in Health Science, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Adriana B. L. Fonseca
- Microcephaly Clinic, Pediatric Division, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Ana J. B. Bispo
- Microcephaly Clinic, Pediatric Division, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Roseane L. S. Porto
- Microcephaly Clinic, Pediatric Division, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Kalliopi Bokea
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Amélia Maria Ribeiro de Jesus
- Molecular Biology Laboratory, Department of Medicine, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Roque Pacheco de Almeida
- Molecular Biology Laboratory, Department of Medicine, University Hospital of the Federal University of Sergipe, Aracaju, Brazil
| | - Rosemary J. Boyton
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Daniel M. Altmann
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
41
|
Luo S, Zhao W, Ma X, Zhang P, Liu B, Zhang L, Wang W, Wang Y, Fu Y, Allain JP, Li T, Li C. A high infectious simian adenovirus type 23 vector based vaccine efficiently protects common marmosets against Zika virus infection. PLoS Negl Trop Dis 2020; 14:e0008027. [PMID: 32049958 PMCID: PMC7015313 DOI: 10.1371/journal.pntd.0008027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 01/03/2020] [Indexed: 01/08/2023] Open
Abstract
Zika virus (ZIKV) has spread in many countries or territories causing severe neurologic complications with potential fatal outcomes. The small primate common marmosets are susceptible to ZIKV, mimicking key features of human infection. Here, a novel simian adenovirus type 23 vector-based vaccine expressing ZIKV pre-membrane-envelope proteins (Sad23L-prM-E) was produced in high infectious titer. Due to determination of immunogenicity in mice, a single-dose of 3×108 PFU Sad23L-prM-E vaccine was intramuscularly inoculated to marmosets. This vaccine raised antibody titers of 104.07 E-specific and 103.13 neutralizing antibody (NAb), as well as robust specific IFN-γ secreting T-cell response (1,219 SFCs/106 cells) to E peptides. The vaccinated marmosets, upon challenge with a high dose of ZIKV (105 PFU) six weeks post prime immunization, reduced viremia by more than 100 folds, and the low level of detectable viral RNA (<103 copies/ml) in blood, saliva, urine and feces was promptly eliminated when the secondary NAb (titer >103.66) and T-cell response (>726 SFCs/106 PBMCs) were acquired 1–2 weeks post exposure to ZIKV, while non-vaccinated control marmosets developed long-term high titer of ZIKV (105.73 copies/ml) (P<0.05). No significant pathological lesions were observed in marmoset tissues. Sad23L-prM-E vaccine was detectable in spleen, liver and PBMCs at least 4 months post challenge. In conclusion, a prime immunization with Sad23L-prM-E vaccine was able to protect marmosets against ZIKV infection when exposed to a high dose of ZIKV. This Sad23L-prM-E vaccine is a promising vaccine candidate for prevention of ZIKV infection in humans. Zika virus (ZIKV) is a member of the Flaviviridaefamily) and causes severe neurologic diseases. The development of safe and effective vaccine is urgent need. In this study, we constructed a novel simian adenovirus type 23 vector-based vaccine expressing ZIKV pre-membrane-envelope proteins (Sad23L-prM-E). By vaccinating the common marmosets with prime immunization of vaccine, and upon challenge with a high dose of ZIKV to the vaccinated marmosets, the immune response and protective efficacy of vaccine were extensively evaluated. The data suggested that Sad23L-prM-E vaccine could protect marmosets against a high dose of ZIKV challenge, which provided a promising vaccine for preventing ZIKV infection in humans.
Collapse
Affiliation(s)
- Shengxue Luo
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Laboratory of Biosafety, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaorui Ma
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Panli Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Bochao Liu
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wenjing Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yuanzhan Wang
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | - Jean-Pierre Allain
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Emeritus professor, University of Cambridge, Cambridge, United Kingdom
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- * E-mail: (TL); (CL)
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- * E-mail: (TL); (CL)
| |
Collapse
|
42
|
Grubor-Bauk B, Wijesundara DK, Masavuli M, Abbink P, Peterson RL, Prow NA, Larocca RA, Mekonnen ZA, Shrestha A, Eyre NS, Beard MR, Gummow J, Carr J, Robertson SA, Hayball JD, Barouch DH, Gowans EJ. NS1 DNA vaccination protects against Zika infection through T cell-mediated immunity in immunocompetent mice. SCIENCE ADVANCES 2019; 5:eaax2388. [PMID: 31844662 PMCID: PMC6905874 DOI: 10.1126/sciadv.aax2388] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/08/2019] [Indexed: 05/08/2023]
Abstract
The causal association of Zika virus (ZIKV) with microcephaly, congenital malformations in infants, and Guillain-Barré syndrome in adults highlights the need for effective vaccines. Thus far, efforts to develop ZIKV vaccines have focused on the viral envelope. ZIKV NS1 as a vaccine immunogen has not been fully explored, although it can circumvent the risk of antibody-dependent enhancement of ZIKV infection, associated with envelope antibodies. Here, we describe a novel DNA vaccine encoding a secreted ZIKV NS1, that confers rapid protection from systemic ZIKV infection in immunocompetent mice. We identify novel NS1 T cell epitopes in vivo and show that functional NS1-specific T cell responses are critical for protection against ZIKV infection. We demonstrate that vaccine-induced anti-NS1 antibodies fail to confer protection in the absence of a functional T cell response. This highlights the importance of using NS1 as a target for T cell-based ZIKV vaccines.
Collapse
Affiliation(s)
- B. Grubor-Bauk
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
- Corresponding author.
| | - D. K. Wijesundara
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - M. Masavuli
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - P. Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - R. L. Peterson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - N. A. Prow
- Experimental Therapeutics Laboratory, Cancer Research Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - R. A. Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Z. A. Mekonnen
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - A. Shrestha
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - N. S. Eyre
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - M. R. Beard
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. Gummow
- Gene Silencing and Expression Core Facility, Adelaide Health and Medical Sciences, Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. Carr
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - S. A. Robertson
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. D. Hayball
- Experimental Therapeutics Laboratory, Cancer Research Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - D. H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - E. J. Gowans
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| |
Collapse
|
43
|
Li G, Adam A, Luo H, Shan C, Cao Z, Fontes-Garfias CR, Sarathy VV, Teleki C, Winkelmann ER, Liang Y, Sun J, Bourne N, Barrett ADT, Shi PY, Wang T. An attenuated Zika virus NS4B protein mutant is a potent inducer of antiviral immune responses. NPJ Vaccines 2019; 4:48. [PMID: 31815005 PMCID: PMC6883050 DOI: 10.1038/s41541-019-0143-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
Live attenuated vaccines (LAVs) are one of the most important strategies to control flavivirus diseases. The flavivirus nonstructural (NS) 4B proteins are a critical component of both the virus replication complex and evasion of host innate immunity. Here we have used site-directed mutagenesis of residues in the highly conserved N-terminal and central hydrophobic regions of Zika virus (ZIKV) NS4B protein to identify candidate attenuating mutations. Three single-site mutants were generated, of which the NS4B-C100S mutant was more attenuated than the other two mutants (NS4B-C100A and NS4B-P36A) in two immunocompromised mouse models of fatal ZIKV disease. The ZIKV NS4B-C100S mutant triggered stronger type 1 interferons and interleukin-6 production, and higher ZIKV-specific CD4+ and CD8+ T-cell responses, but induced similar titers of neutralization antibodies compared with the parent wild-type ZIKV strain and a previously reported candidate ZIKV LAV with a 10-nucleotide deletion in 3'-UTR (ZIKV-3'UTR-Δ10). Vaccination with ZIKV NS4B-C100S protected mice from subsequent WT ZIKV challenge. Furthermore, either passive immunization with ZIKV NS4B-C100S immune sera or active immunization with ZIKV NS4B-C100S followed by the depletion of T cells affords full protection from lethal WT ZIKV challenge. In summary, our results suggest that the ZIKV NS4B-C100S mutant may serve as a candidate ZIKV LAV due to its attenuated phenotype and high immunogenicity.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Awadalkareem Adam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Huanle Luo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Chao Shan
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Zengguo Cao
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Camila R. Fontes-Garfias
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Vanessa V. Sarathy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Cody Teleki
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Evandro R. Winkelmann
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Nigel Bourne
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Alan D. T. Barrett
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Tian Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
| |
Collapse
|
44
|
Effects of Adjuvants on the Immunogenicity and Efficacy of a Zika Virus Envelope Domain III Subunit Vaccine. Vaccines (Basel) 2019; 7:vaccines7040161. [PMID: 31717890 PMCID: PMC6963592 DOI: 10.3390/vaccines7040161] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/10/2019] [Accepted: 10/24/2019] [Indexed: 02/08/2023] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, has attracted global attention due to its close association with congenital Zika syndrome and neurological diseases, and transmission through additional routes, such as sexual contact. Currently there are no vaccines approved for ZIKV, and thus, there is an urgent need to develop an effective and safe ZIKV vaccine. Domain III (DIII) of the ZIKV envelope (E) protein is an important vaccine target, and a vaccine developed using a mutant DIII of E (EDIII) protein protects adult and pregnant mice, and unborn offspring, against ZIKV infection. Here, we have used immunocompetent BALB/c mice treated with anti-interferon-α/β receptor 1 (Ifnar1) antibodies to investigate whether three adjuvants (aluminum (Alum), monophosphoryl lipid A (MPL), and MF59), either alone or in combination, could improve the efficacy of this EDIII subunit vaccine. Our data show that, although vaccine formulated with a single adjuvant induced a specific antibody and cellular immune response, and reduced viral load in mice challenged with ZIKV, the combination of Alum and MPL adjuvants led to a more robust and balanced immune response, stronger neutralizing activity against three recent ZIKV human strains, and greater protection against a high-dose ZIKV challenge. Particularly, the combination of Alum with MPL significantly reduced viral titers and viral RNA copy numbers in sera and tissues, including the male reproductive organs. Overall, this study has identified the combination of Alum and MPL as the most effective adjuvant for ZIKV EDIII subunit vaccines, and it has important implications for subunit vaccines against other enveloped viruses, including non-ZIKV flaviviruses.
Collapse
|
45
|
Pérez-Guzmán EX, Pantoja P, Serrano-Collazo C, Hassert MA, Ortiz-Rosa A, Rodríguez IV, Giavedoni L, Hodara V, Parodi L, Cruz L, Arana T, White LJ, Martínez MI, Weiskopf D, Brien JD, de Silva A, Pinto AK, Sariol CA. Time elapsed between Zika and dengue virus infections affects antibody and T cell responses. Nat Commun 2019; 10:4316. [PMID: 31541110 PMCID: PMC6754404 DOI: 10.1038/s41467-019-12295-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are co-endemic in many parts of the world, but the impact of ZIKV infection on subsequent DENV infection is not well understood. Here we show in rhesus macaques that the time elapsed after ZIKV infection affects the immune response to DENV infection. We show that previous ZIKV exposure increases the magnitude of the antibody and T cell responses against DENV. The time interval between ZIKV and subsequent DENV infection further affects the immune response. A mid-convalescent period of 10 months after ZIKV infection results in higher and more durable antibody and T cell responses to DENV infection than a short period of 2 months. In contrast, previous ZIKV infection does not affect DENV viremia or pro-inflammatory status. Collectively, we find no evidence of a detrimental effect of ZIKV immunity in a subsequent DENV infection. This supports the implementation of ZIKV vaccines that could also boost immunity against future DENV epidemics.
Collapse
Affiliation(s)
- Erick X Pérez-Guzmán
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
- Takeda Vaccines Inc, Cambridge, MA, USA
| | - Petraleigh Pantoja
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Crisanta Serrano-Collazo
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Mariah A Hassert
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Alexandra Ortiz-Rosa
- Department of Biology, University of Puerto Rico-Río Piedras Campus, San Juan, PR, USA
| | - Idia V Rodríguez
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Luis Giavedoni
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Vida Hodara
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Laura Parodi
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Lorna Cruz
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Teresa Arana
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Laura J White
- Departments of Microbiology & Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Melween I Martínez
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - James D Brien
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Aravinda de Silva
- Departments of Microbiology & Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Amelia K Pinto
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Carlos A Sariol
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA.
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA.
- Department of Internal Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, 00936, PR, USA.
| |
Collapse
|
46
|
Vδ2 T-Cells Kill ZIKV-Infected Cells by NKG2D-Mediated Cytotoxicity. Microorganisms 2019; 7:microorganisms7090350. [PMID: 31547470 PMCID: PMC6781265 DOI: 10.3390/microorganisms7090350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/23/2022] Open
Abstract
An expansion of effector/activated Vδ2 T-cells was recently described in acute Zika virus (ZIKV)-infected patients, but their role in the protective immune response was not clarified. The aim of this study was to define the antiviral activity of Vδ2 T-cells against ZIKV-infected cells. The Vδ2 T-cells expansion and their cytotoxic activity against ZIKV-infected cells were tested in vitro and analyzed by RT-PCR and flow cytometry. We found that ZIKV infection was able to induce Vδ2 T-cells expansion and sensitized A549 cells to Vδ2-mediated killing. Indeed, expanded Vδ2 T-cells killed ZIKV-infected cells through degranulation and perforin release. Moreover, ZIKV infection was able to increase the expression on A549 cells of NKG2D ligands (NKG2DLs), namely MICA, MICB, and ULBP2, at both the mRNA and protein levels, suggesting the possible involvement of these molecules in the recognition by NKG2D-expressing Vδ2 T-cells. Indeed, the killing of ZIKV-infected cells by expanded Vδ2 T-cells was mediated by NKG2D/NKG2DL interaction as NKG2D neutralization abrogated Vδ2 cytotoxicity. Our data showed a strong antiviral activity of Vδ2 T-cells against ZIKV-infected cells, suggesting their involvement in the protective immune response. Other studies are necessary to investigate whether the lack of Vδ2 T-cells expansion in vivo may be associated with disease complications.
Collapse
|
47
|
Pardy RD, Richer MJ. Protective to a T: The Role of T Cells during Zika Virus Infection. Cells 2019; 8:cells8080820. [PMID: 31382545 PMCID: PMC6721718 DOI: 10.3390/cells8080820] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/22/2022] Open
Abstract
CD4 and CD8 T cells are an important part of the host's capacity to defend itself against viral infections. During flavivirus infections, T cells have been implicated in both protective and pathogenic responses. Given the recent emergence of Zika virus (ZIKV) as a prominent global health threat, the question remains as to how T cells contribute to anti-ZIKV immunity. Furthermore, high homology between ZIKV and other, co-circulating flaviviruses opens the possibility of positive or negative effects of cross-reactivity due to pre-existing immunity. In this review, we will discuss the CD4 and CD8 T cell responses to ZIKV, and the lessons we have learned from both mouse and human infections. In addition, we will consider the possibility of whether T cells, in the context of flavivirus-naïve and flavivirus-immune subjects, play a role in promoting ZIKV pathogenesis during infection.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Martin J Richer
- Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 2B4, Canada.
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
48
|
Branche E, Simon AY, Sheets N, Kim K, Barker D, Nguyen AVT, Sahota H, Young MP, Salgado R, Mamidi A, Viramontes KM, Carnelley T, Qiu H, Elong Ngono A, Regla-Nava JA, Susantono MX, Valls Cuevas JM, Kennedy K, Kodihalli S, Shresta S. Human Polyclonal Antibodies Prevent Lethal Zika Virus Infection in Mice. Sci Rep 2019; 9:9857. [PMID: 31285451 PMCID: PMC6614477 DOI: 10.1038/s41598-019-46291-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 06/17/2019] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that represents a major threat to global health. ZIKV infections in adults are generally asymptomatic or present with mild symptoms. However, recent outbreaks of ZIKV have revealed that it can cause Congenital Zika Syndrome in neonates and Guillain-Barré syndrome in adults. Currently, no ZIKV-specific vaccines or antiviral treatments are available. In this study, we tested the efficacy of convalescent plasma IgG hyperimmune product (ZIKV-IG) isolated from individuals with high neutralizing anti-ZIKV titers as a therapeutic candidate against ZIKV infection using a model of ZIKV infection in Ifnar1-/- mice. ZIKV-IG successfully protected mice from lethal ZIKV challenge. In particular, ZIKV-IG treatment at 24 hours after lethal ZIKV infection improved survival by reducing weight loss and tissue viral burden and improving clinical score. Additionally, ZIKV-IG eliminated ZIKV-induced tissue damage and inflammation in the brain and liver. These results indicate that ZIKV-IG is efficacious against ZIKV, suggesting this human polyclonal antibody is a viable candidate for further development as a treatment against human ZIKV infection.
Collapse
Affiliation(s)
- Emilie Branche
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Ayo Yila Simon
- Research and Development, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Nicholas Sheets
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Douglas Barker
- Research and Development, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Anh-Viet T Nguyen
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Harpreet Sahota
- Medical Affairs, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Matthew Perry Young
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Rebecca Salgado
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Anila Mamidi
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Karla M Viramontes
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Trevor Carnelley
- Research and Development, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Hongyu Qiu
- Research and Development, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Annie Elong Ngono
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | | | | | - Joan M Valls Cuevas
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Kieron Kennedy
- Research and Development, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Shantha Kodihalli
- Research and Development, Emergent BioSolutions Canada Inc, 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada.
| | - Sujan Shresta
- La Jolla Institute for Immunology 9420 Athena Circle, La Jolla, CA, 92037, USA.
| |
Collapse
|
49
|
Elong Ngono A, Young MP, Bunz M, Xu Z, Hattakam S, Vizcarra E, Regla-Nava JA, Tang WW, Yamabhai M, Wen J, Shresta S. Correction: CD4+ T cells promote humoral immunity and viral control during Zika virus infection. PLoS Pathog 2019; 15:e1007821. [PMID: 31136625 PMCID: PMC6538173 DOI: 10.1371/journal.ppat.1007821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|