1
|
Yadav N, Kalata AC, Reynolds RA, Raappana A, Sather DN, Murphy SC. Identifying Plasmodium P36 and P52 antigens for coadministration with circumsporozoite protein to enhance vaccine efficacy. NPJ Vaccines 2024; 9:241. [PMID: 39643623 PMCID: PMC11624287 DOI: 10.1038/s41541-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024] Open
Abstract
Vaccines targeting the complex pre-erythrocytic stage of Plasmodium parasites may benefit from the inclusion of multiple antigens. However, discerning protective effects can be difficult because newer candidates may not be as protective as leading antigens like the circumsporozoite protein (CSP) in the conventional pre-clinical mouse model. We developed a modified mouse model challenge strategy that maximizes the contribution of T cells induced by novel candidate antigens at the sporozoite challenge time point and used this approach to test Plasmodium P36 and P52 vaccine candidates alone and in concert with non-protective doses of CSP. Co-administration of P36 and/or P52 with CSP achieved 80-100% sterile protection in mice, compared to only 7-30% protection for each individual antigen. P36 and P52 vaccination induced murine CD4+ and CD8+ T cell responses, but not antibody responses. This study adds P36 and P52 as promising vaccine antigens that may enhance the protection achieved by CSP vaccination.
Collapse
Affiliation(s)
- Naveen Yadav
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Anya C Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Rebekah A Reynolds
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, USA
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA.
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA.
- Department of Microbiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Yadav N, Kalata AC, Reynolds RA, Raappana A, Sather DN, Murphy SC. Identifying Plasmodium P36 and P52 antigens for co-administration with circumsporozoite protein to enhance vaccine efficacy. RESEARCH SQUARE 2024:rs.3.rs-4909396. [PMID: 39399676 PMCID: PMC11469399 DOI: 10.21203/rs.3.rs-4909396/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Vaccines targeting the complex pre-erythrocytic stage of Plasmodium parasites may benefit from inclusion of multiple antigens. However, discerning protective effects can be difficult because newer candidates may not be as protective as leading antigens like the circumsporozoite protein (CSP) in the conventional pre-clinical mouse model. We developed a modified mouse model challenge strategy that maximizes the contribution of T cells induced by novel candidate antigens at the sporozoite challenge time point and used this approach to test Plasmodium P36 and P52 vaccine candidates alone and in concert with non-protective doses of CSP. Co-administration of P36 and/or P52 with CSP achieved 80-100% sterile protection in mice, compared to only 7-30% protection for each individual antigen. P36 and P52 vaccination induced murine CD4+ and CD8+ T cell responses, but not antibody responses. This study adds P36 and P52 as promising vaccine antigens that may enhance protection achieved by CSP vaccination.
Collapse
Affiliation(s)
- Naveen Yadav
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Anya C. Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Rebekah A. Reynolds
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Andrew Raappana
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - D. Noah Sather
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| |
Collapse
|
3
|
Xu Y, Sun F, Bai Z, Bian C, Wang X, Zhao Z, Yang P. Cold-adapted influenza-vectored RSV vaccine protects BALB/c mice and cotton rats from RSV challenge. J Med Virol 2024; 96:e29308. [PMID: 39007405 DOI: 10.1002/jmv.29308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 11/03/2023] [Accepted: 11/27/2023] [Indexed: 07/16/2024]
Abstract
Respiratory syncytial virus (RSV) remains the primary cause of lower respiratory tract infections, particularly in infants and the elderly. In this study, we employed reverse genetics to generate a chimeric influenza virus expressing neuraminidase-3F protein conjugate with three repeats of the RSV F protein protective epitope inserted into the NA gene of A/California/7/2009 ca (CA/AA ca), resulting in rFlu/RSV/NA-3F (hereafter, rFRN3). The expression of NA-3F protein was confirmed by Western blotting. The morphology and temperature-sensitive phenotype of rFRN3 were similar to CA/AA ca. Its immunogenicity and protective efficiency were evaluated in BALB/c mice and cotton rats. Intranasal administration of rFRN3 elicited robust humoral, cellular, and to some extent, mucosal immune responses. Compared to controls, rFRN3 protected animals from RSV infection, attenuated lung injury, and reduced viral titers in the nose and lungs post-RSV challenge. These results demonstrate that rFRN3 can trigger RSV-specific immune responses and thus exhibits potent protective efficacy. The "dual vaccine" approach of a cold-adapted influenza vector RSV vaccine will improve the prophylaxis of influenza and RSV infection. rFRN3 thus warrants further clinical investigations as a candidate RSV vaccine.
Collapse
Affiliation(s)
- Yongru Xu
- The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
| | - Fang Sun
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhifang Bai
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chengrong Bian
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Penghui Yang
- The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
| |
Collapse
|
4
|
Meseko C, Sanicas M, Asha K, Sulaiman L, Kumar B. Antiviral options and therapeutics against influenza: history, latest developments and future prospects. Front Cell Infect Microbiol 2023; 13:1269344. [PMID: 38094741 PMCID: PMC10716471 DOI: 10.3389/fcimb.2023.1269344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Drugs and chemotherapeutics have helped to manage devastating impacts of infectious diseases since the concept of 'magic bullet'. The World Health Organization estimates about 650,000 deaths due to respiratory diseases linked to seasonal influenza each year. Pandemic influenza, on the other hand, is the most feared health disaster and probably would have greater and immediate impact on humanity than climate change. While countermeasures, biosecurity and vaccination remain the most effective preventive strategies against this highly infectious and communicable disease, antivirals are nonetheless essential to mitigate clinical manifestations following infection and to reduce devastating complications and mortality. Continuous emergence of the novel strains of rapidly evolving influenza viruses, some of which are intractable, require new approaches towards influenza chemotherapeutics including optimization of existing anti-infectives and search for novel therapies. Effective management of influenza infections depend on the safety and efficacy of selected anti-infective in-vitro studies and their clinical applications. The outcomes of therapies are also dependent on understanding diversity in patient groups, co-morbidities, co-infections and combination therapies. In this extensive review, we have discussed the challenges of influenza epidemics and pandemics and discoursed the options for anti-viral chemotherapies for effective management of influenza virus infections.
Collapse
Affiliation(s)
- Clement Meseko
- Regional Centre for Animal Influenza, National Veterinary Research Institute, Vom, Nigeria
| | - Melvin Sanicas
- Medical and Clinical Development, Clover Biopharmaceuticals, Boston, MA, United States
| | - Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Lanre Sulaiman
- Regional Centre for Animal Influenza, National Veterinary Research Institute, Vom, Nigeria
| | - Binod Kumar
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
5
|
Beicht J, Kubinski M, Zdora I, Puff C, Biermann J, Gerlach T, Baumgärtner W, Sutter G, Osterhaus ADME, Prajeeth CK, Rimmelzwaan GF. Induction of humoral and cell-mediated immunity to the NS1 protein of TBEV with recombinant Influenza virus and MVA affords partial protection against lethal TBEV infection in mice. Front Immunol 2023; 14:1177324. [PMID: 37483628 PMCID: PMC10360051 DOI: 10.3389/fimmu.2023.1177324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Tick-borne encephalitis virus (TBEV) is one of the most relevant tick-transmitted neurotropic arboviruses in Europe and Asia and the causative agent of tick-borne encephalitis (TBE). Annually more than 10,000 TBE cases are reported despite having vaccines available. In Europe, the vaccines FSME-IMMUN® and Encepur® based on formaldehyde-inactivated whole viruses are licensed. However, demanding vaccination schedules contribute to sub-optimal vaccination uptake and breakthrough infections have been reported repeatedly. Due to its immunogenic properties as well as its role in viral replication and disease pathogenesis, the non-structural protein 1 (NS1) of flaviviruses has become of interest for non-virion based flavivirus vaccine candidates in recent years. Methods Therefore, immunogenicity and protective efficacy of TBEV NS1 expressed by neuraminidase (NA)-deficient Influenza A virus (IAV) or Modified Vaccinia virus Ankara (MVA) vectors were investigated in this study. Results With these recombinant viral vectors TBEV NS1-specific antibody and T cell responses were induced. Upon heterologous prime/boost regimens partial protection against lethal TBEV challenge infection was afforded in mice. Discussion This supports the inclusion of NS1 as a vaccine component in next generation TBEV vaccines.
Collapse
Affiliation(s)
- Jana Beicht
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Center for Systems Neuroscience, Hannover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hannover, Germany
| | - Christina Puff
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Jeannine Biermann
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Center for Systems Neuroscience, Hannover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Albert D. M. E. Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
6
|
Hassert M, Arumugam S, Harty JT. Memory CD8+ T cell-mediated protection against liver-stage malaria. Immunol Rev 2023; 316:84-103. [PMID: 37014087 PMCID: PMC10524177 DOI: 10.1111/imr.13202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/09/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Nearly half of the world's population is at risk of malaria, a disease caused by the protozoan parasite Plasmodium, which is estimated to cause more than 240,000,000 infections and kill more than 600,000 people annually. The emergence of Plasmodia resistant to chemoprophylactic treatment highlights the urgency to develop more effective vaccines. In this regard, whole sporozoite vaccination approaches in murine models and human challenge studies have provided substantial insight into the immune correlates of protection from malaria. From these studies, CD8+ T cells have come to the forefront, being identified as critical for vaccine-mediated liver-stage immunity that can prevent the establishment of the symptomatic blood stages and subsequent transmission of infection. However, the unique biological characteristics required for CD8+ T cell protection from liver-stage malaria dictate that more work must be done to design effective vaccines. In this review, we will highlight a subset of studies that reveal basic aspects of memory CD8+ T cell-mediated protection from liver-stage malaria infection.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| | - Sahaana Arumugam
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| | - John T. Harty
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
7
|
O'Connor JH, McNamara HA, Cai Y, Coupland LA, Gardiner EE, Parish CR, McMorran BJ, Ganusov VV, Cockburn IA. Interactions with Asialo-Glycoprotein Receptors and Platelets Are Dispensable for CD8 + T Cell Localization in the Murine Liver. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2738-2748. [PMID: 35649630 PMCID: PMC9308657 DOI: 10.4049/jimmunol.2101037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
Liver-resident CD8+ T cells can play critical roles in the control of pathogens, including Plasmodium and hepatitis B virus. Paradoxically, it has also been proposed that the liver may act as the main place for the elimination of CD8+ T cells at the resolution of immune responses. We hypothesized that different adhesion processes may drive residence versus elimination of T cells in the liver. Specifically, we investigated whether the expression of asialo-glycoproteins (ASGPs) drives the localization and elimination of effector CD8+ T cells in the liver, while interactions with platelets facilitate liver residence and protective function. Using murine CD8+ T cells activated in vitro, or in vivo by immunization with Plasmodium berghei sporozoites, we found that, unexpectedly, inhibition of ASGP receptors did not inhibit the accumulation of effector cells in the liver, but instead prevented these cells from accumulating in the spleen. In addition, enforced expression of ASGP on effector CD8+ T cells using St3GalI-deficient cells lead to their loss from the spleen. We also found, using different mouse models of thrombocytopenia, that severe reduction in platelet concentration in circulation did not strongly influence the residence and protective function of CD8+ T cells in the liver. These data suggest that platelets play a marginal role in CD8+ T cell function in the liver. Furthermore, ASGP-expressing effector CD8+ T cells accumulate in the spleen, not the liver, prior to their destruction.
Collapse
Affiliation(s)
- James H O'Connor
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- Australian National University Medical School, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Hayley A McNamara
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Yeping Cai
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Lucy A Coupland
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia; and
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia; and
| | - Christopher R Parish
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia; and
| | - Brendan J McMorran
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Vitaly V Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN
| | - Ian A Cockburn
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia;
| |
Collapse
|
8
|
Mooij P, García-Arriaza J, Pérez P, Lázaro-Frías A, Verstrepen BE, Böszörményi KP, Mortier D, Fagrouch Z, Kiemenyi-Kayere G, Niphuis H, Acar RF, Meijer L, Stammes MA, Kondova I, Verschoor EJ, GeurtsvanKessel CH, de Bruin E, Sikkema RS, Luczkowiak J, Delgado R, Montenegro D, Puentes E, Rodríguez E, Bogers WMJM, Koopman G, Esteban M. Poxvirus MVA Expressing SARS-CoV-2 S Protein Induces Robust Immunity and Protects Rhesus Macaques From SARS-CoV-2. Front Immunol 2022; 13:845887. [PMID: 35371043 PMCID: PMC8966779 DOI: 10.3389/fimmu.2022.845887] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
Novel safe, immunogenic, and effective vaccines are needed to control the COVID-19 pandemic, caused by SARS-CoV-2. Here, we describe the safety, robust immunogenicity, and potent efficacy elicited in rhesus macaques by a modified vaccinia virus Ankara (MVA) vector expressing a full-length SARS-CoV-2 spike (S) protein (MVA-S). MVA-S vaccination was well tolerated and induced S and receptor-binding domain (RBD)-binding IgG antibodies and neutralizing antibodies against SARS-CoV-2 and several variants of concern. S-specific IFNγ, but not IL-4, -producing cells were also elicited. After SARS-CoV-2 challenge, vaccinated animals showed a significant strong reduction of virus loads in bronchoalveolar lavages (BAL) and decreased levels in throat and nasal mucosa. Remarkably, MVA-S also protected macaques from fever and infection-induced cytokine storm. Computed tomography and histological examination of the lungs showed reduced lung pathology in MVA-S-vaccinated animals. These findings favor the use of MVA-S as a potential vaccine for SARS-CoV-2 in clinical trials.
Collapse
Affiliation(s)
- Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Adrian Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Babs E. Verstrepen
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Kinga P. Böszörményi
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | | | - Henk Niphuis
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Roja Fidel Acar
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Lisette Meijer
- Department of Parasitology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Marieke A. Stammes
- Department of Parasitology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Ivanela Kondova
- Animal Science Department, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Ernst J. Verschoor
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | | | - Erwin de Bruin
- Department of Viroscience, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Reina S. Sikkema
- Department of Viroscience, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense School of Medicine, Madrid, Spain
| | | | | | | | - Willy M. J. M. Bogers
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
9
|
Lázaro-Frías A, Pérez P, Zamora C, Sánchez-Cordón PJ, Guzmán M, Luczkowiak J, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. Full efficacy and long-term immunogenicity induced by the SARS-CoV-2 vaccine candidate MVA-CoV2-S in mice. NPJ Vaccines 2022; 7:17. [PMID: 35140227 PMCID: PMC8828760 DOI: 10.1038/s41541-022-00440-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
Two doses of the MVA-CoV2-S vaccine candidate expressing the SARS-CoV-2 spike (S) protein protected K18-hACE2 transgenic mice from a lethal dose of SARS-CoV-2. This vaccination regimen prevented virus replication in the lungs, reduced lung pathology, and diminished levels of pro-inflammatory cytokines. High titers of IgG antibodies against S and receptor-binding domain (RBD) proteins and of neutralizing antibodies were induced against parental virus and variants of concern, markers that correlated with protection. Similar SARS-CoV-2-specific antibody responses were observed at prechallenge and postchallenge in the two-dose regimen, while the single-dose treatment does not avoid vaccine breakthrough infection. All vaccinated animals survived infection and were also protected to SARS-CoV-2 reinfection. Furthermore, two MVA-CoV2-S doses induced long-term memory S-specific humoral and cellular immune responses in C57BL/6 mice, 6 months after immunization. The efficacy and immunological benefits of the MVA-CoV2-S vaccine candidate against COVID-19 supports its consideration for human clinical trials.
Collapse
Affiliation(s)
- Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain
| | - Pedro J Sánchez-Cordón
- Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), 28130, Valdeolmos, Madrid, Spain
| | - María Guzmán
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), 28041, Madrid, Spain.,Universidad Complutense School of Medicine, 28040, Madrid, Spain
| | - José M Casasnovas
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain.
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| |
Collapse
|
10
|
Pérez P, Martín-Acebes MA, Poderoso T, Lázaro-Frías A, Saiz JC, Sorzano CÓS, Esteban M, García-Arriaza J. The combined vaccination protocol of DNA/MVA expressing Zika virus structural proteins as efficient inducer of T and B cell immune responses. Emerg Microbes Infect 2021; 10:1441-1456. [PMID: 34213405 PMCID: PMC8284158 DOI: 10.1080/22221751.2021.1951624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen with public health importance due to the high risk of its mosquito vector dissemination and the severe neurological and teratogenic sequelae associated with infection. Vaccines with broad immune specificity and control against this re-emerging virus are needed. Here, we described that mice immunized with a priming dose of a DNA plasmid mammalian expression vector encoding ZIKV prM-E antigens (DNA-ZIKV) followed by a booster dose of a modified vaccinia virus Ankara (MVA) vector expressing the same prM-E ZIKV antigens (MVA-ZIKV) induced broad, polyfunctional and long-lasting ZIKV-specific CD4+ and CD8+ T-cell immune responses, with high levels of CD4+ T follicular helper cells, together with the induction of neutralizing antibodies. All those immune parameters were significantly stronger in the heterologous DNA-ZIKV/MVA-ZIKV immunization group compared to the homologous prime/boost immunizations regimens. Collectively, these results provided an optimized immunization protocol able to induce high levels of ZIKV-specific T-cell responses, as well as neutralizing antibodies and reinforce the combined use of DNA-based vectors and MVA-ZIKV as promising prophylactic vaccination schedule against ZIKV.
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Miguel A. Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Teresa Poderoso
- Molecular Virology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carlos Óscar S. Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain, Mariano Esteban
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain,Juan García-Arriaza
| |
Collapse
|
11
|
Pirahmadi S, Afzali S, Zargar M, Zakeri S, Mehrizi AA. How can we develop an effective subunit vaccine to achieve successful malaria eradication? Microb Pathog 2021; 160:105203. [PMID: 34547408 DOI: 10.1016/j.micpath.2021.105203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/05/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022]
Abstract
Malaria, a mosquito-borne infection, is the most widespread parasitic disease. Despite numerous efforts to eradicate malaria, this disease is still a health concern worldwide. Owing to insecticide-resistant vectors and drug-resistant parasites, available controlling measures are insufficient to achieve a malaria-free world. Thus, there is an urgent need for new intervention tools such as efficient malaria vaccines. Subunit vaccines are the most promising malaria vaccines under development. However, one of the major drawbacks of subunit vaccines is the lack of efficient and durable immune responses including antigen-specific antibody, CD4+, and CD8+ T-cell responses, long-lived plasma cells, memory cells, and functional antibodies for parasite neutralization or inhibition of parasite invasion. These types of responses could be induced by whole organism vaccines, but eliciting these responses with subunit vaccines has been proven to be more challenging. Consequently, subunit vaccines require several policies to overcome these challenges. In this review, we address common approaches that can improve the efficacy of subunit vaccines against malaria.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shima Afzali
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Zargar
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
12
|
Gómez CE, Perdiguero B, Usero L, Marcos-Villar L, Miralles L, Leal L, Sorzano CÓS, Sánchez-Corzo C, Plana M, García F, Esteban M. Enhancement of the HIV-1-Specific Immune Response Induced by an mRNA Vaccine through Boosting with a Poxvirus MVA Vector Expressing the Same Antigen. Vaccines (Basel) 2021; 9:vaccines9090959. [PMID: 34579196 PMCID: PMC8473054 DOI: 10.3390/vaccines9090959] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
Development of a vaccine against HIV remains a major target goal in the field. The recent success of mRNA vaccines against the coronavirus SARS-CoV-2 is pointing out a new era of vaccine designs against pathogens. Here, we have generated two types of mRNA vaccine candidates against HIV-1; one based on unmodified vectors and the other on 1-methyl-3′-pseudouridylyl modified vectors expressing a T cell multiepitopic construct including protective conserved epitopes from HIV-1 Gag, Pol and Nef proteins (referred to as RNA-TMEP and RNA-TMEPmod, respectively) and defined their biological and immunological properties in cultured cells and in mice. In cultured cells, both mRNA vectors expressed the corresponding protein, with higher levels observed in the unmodified mRNA, leading to activated macrophages with differential induction of innate immune molecules. In mice, intranodal administration of the mRNAs induced the activation of specific T cell (CD4 and CD8) responses, and the levels were markedly enhanced after a booster immunization with the poxvirus vector MVA-TMEP expressing the same antigen. This immune activation was maintained even three months later. These findings revealed a potent combined immunization regimen able to enhance the HIV-1-specific immune responses induced by an mRNA vaccine that might be applicable to human vaccination programs with mRNA and MVA vectors.
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Centro Nacional de Biotecnología (CNB), Department of Molecular and Cellular Biology, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (B.P.); (L.M.-V.); (C.S.-C.)
- Correspondence: (C.E.G.); (M.E.)
| | - Beatriz Perdiguero
- Centro Nacional de Biotecnología (CNB), Department of Molecular and Cellular Biology, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (B.P.); (L.M.-V.); (C.S.-C.)
| | - Lorena Usero
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (L.U.); (L.M.); (L.L.); (M.P.); (F.G.)
| | - Laura Marcos-Villar
- Centro Nacional de Biotecnología (CNB), Department of Molecular and Cellular Biology, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (B.P.); (L.M.-V.); (C.S.-C.)
| | - Laia Miralles
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (L.U.); (L.M.); (L.L.); (M.P.); (F.G.)
| | - Lorna Leal
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (L.U.); (L.M.); (L.L.); (M.P.); (F.G.)
| | | | - Cristina Sánchez-Corzo
- Centro Nacional de Biotecnología (CNB), Department of Molecular and Cellular Biology, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (B.P.); (L.M.-V.); (C.S.-C.)
| | - Montserrat Plana
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (L.U.); (L.M.); (L.L.); (M.P.); (F.G.)
| | - Felipe García
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (L.U.); (L.M.); (L.L.); (M.P.); (F.G.)
| | - Mariano Esteban
- Centro Nacional de Biotecnología (CNB), Department of Molecular and Cellular Biology, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (B.P.); (L.M.-V.); (C.S.-C.)
- Correspondence: (C.E.G.); (M.E.)
| |
Collapse
|
13
|
Moraschi BF, Noronha IH, Ferreira CP, Cariste LM, Monteiro CB, Denapoli P, Vrechi T, Pereira GJS, Gazzinelli RT, Lannes-Vieira J, Rodrigues MM, Bortoluci KR, Vasconcelos JRC. Rapamycin Improves the Response of Effector and Memory CD8 + T Cells Induced by Immunization With ASP2 of Trypanosoma cruzi. Front Cell Infect Microbiol 2021; 11:676183. [PMID: 34123875 PMCID: PMC8191465 DOI: 10.3389/fcimb.2021.676183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
Deficiency in memory formation and increased immunosenescence are pivotal features of Trypanosoma cruzi infection proposed to play a role in parasite persistence and disease development. The vaccination protocol that consists in a prime with plasmid DNA followed by the boost with a deficient recombinant human adenovirus type 5, both carrying the ASP2 gene of T. cruzi, is a powerful strategy to elicit effector memory CD8+ T-cells against this parasite. In virus infections, the inhibition of mTOR, a kinase involved in several biological processes, improves the response of memory CD8+ T-cells. Therefore, our aim was to assess the role of rapamycin, the pharmacological inhibitor of mTOR, in CD8+ T response against T. cruzi induced by heterologous prime-boost vaccine. For this purpose, C57BL/6 or A/Sn mice were immunized and daily treated with rapamycin for 34 days. CD8+ T-cells response was evaluated by immunophenotyping, intracellular staining, ELISpot assay and in vivo cytotoxicity. In comparison with vehicle-injection, rapamycin administration during immunization enhanced the frequency of ASP2-specific CD8+ T-cells and the percentage of the polyfunctional population, which degranulated (CD107a+) and secreted both interferon gamma (IFNγ) and tumor necrosis factor (TNF). The beneficial effects were long-lasting and could be detected 95 days after priming. Moreover, the effects were detected in mice immunized with ten-fold lower doses of plasmid/adenovirus. Additionally, the highly susceptible to T. cruzi infection A/Sn mice, when immunized with low vaccine doses, treated with rapamycin, and challenged with trypomastigote forms of the Y strain showed a survival rate of 100%, compared with 42% in vehicle-injected group. Trying to shed light on the biological mechanisms involved in these beneficial effects on CD8+ T-cells by mTOR inhibition after immunization, we showed that in vivo proliferation was higher after rapamycin treatment compared with vehicle-injected group. Taken together, our data provide a new approach to vaccine development against intracellular parasites, placing the mTOR inhibitor rapamycin as an adjuvant to improve effective CD8+ T-cell response.
Collapse
Affiliation(s)
- Barbara Ferri Moraschi
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Isaú Henrique Noronha
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Camila Pontes Ferreira
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Leonardo M. Cariste
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, Santos, Brazil
| | - Caroline B. Monteiro
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, Santos, Brazil
| | - Priscila Denapoli
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Talita Vrechi
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Gustavo J. S. Pereira
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - Ricardo T. Gazzinelli
- René Rachou Research Center, Fiocruz, Belo Horizonte, Brazil
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Joseli Lannes-Vieira
- Laboratoy of Biology of the Interactions, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Maurício M. Rodrigues
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Karina R. Bortoluci
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Department of Pharmacology, Federal University of São Paulo, (UNIFESP), São Paulo, Brazil
| | - José Ronnie C. Vasconcelos
- Molecular Immunology Laboratory, Center of Molecular and Cellular Therapy, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, Santos, Brazil
| |
Collapse
|
14
|
Tiwari S, Dutt TS, Chen B, Chen M, Kim J, Dai AZ, Lukose R, Shanley C, Fox A, Karger BR, Porcelli SA, Chan J, Podell BK, Obregon-Henao A, Orme IM, Jacobs WR, Henao-Tamayo M. BCG-Prime and boost with Esx-5 secretion system deletion mutant leads to better protection against clinical strains of Mycobacterium tuberculosis. Vaccine 2020; 38:7156-7165. [PMID: 32978002 PMCID: PMC7755135 DOI: 10.1016/j.vaccine.2020.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/07/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022]
Abstract
Although vaccination with BCG prevents disseminated forms of childhood tuberculosis (TB), it does not protect against pulmonary infection or Mycobacterium tuberculosis (Mtb) transmission. In this study, we generated a complete deletion mutant of the Mtb Esx-5 type VII secretion system (Mtb Δesx-5). Mtb Δesx-5 was highly attenuated and safe in immunocompromised mice. When tested as a vaccine candidate to boost BCG-primed immunity, Mtb Δesx-5 improved protection against highly virulent Mtb strains in the murine and guinea pig models of TB. Enhanced protection provided by heterologous BCG-prime plus Mtb Δesx-5 boost regimen was associated with increased pulmonary influx of central memory T cells (TCM), follicular helper T cells (TFH) and activated monocytes. Conversely, lower numbers of T cells expressing exhaustion markers were observed in vaccinated animals. Our results suggest that boosting BCG-primed immunity with Mtb Δesx-5 is a potential approach to improve protective immunity against Mtb. Further insight into the mechanism of action of this novel prime-boost approach is warranted.
Collapse
Affiliation(s)
- Sangeeta Tiwari
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, United States.
| | - Taru S Dutt
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Bing Chen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Mei Chen
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, United States; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - John Kim
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Annie Zhi Dai
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Regy Lukose
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Crystal Shanley
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Amy Fox
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Burton R Karger
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - John Chan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Brendan K Podell
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Andres Obregon-Henao
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Ian M Orme
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Marcela Henao-Tamayo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States.
| |
Collapse
|
15
|
Importance of the Immunodominant CD8 + T Cell Epitope of Plasmodium berghei Circumsporozoite Protein in Parasite- and Vaccine-Induced Protection. Infect Immun 2020; 88:IAI.00383-20. [PMID: 32719159 DOI: 10.1128/iai.00383-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The circumsporozoite protein (CSP) builds up the surface coat of sporozoites and is the leading malaria pre-erythrocytic-stage vaccine candidate. CSP has been shown to induce robust CD8+ T cell responses that are capable of eliminating developing parasites in hepatocytes, resulting in protective immunity. In this study, we characterized the importance of the immunodominant CSP-derived epitope SYIPSAEKI of Plasmodium berghei in both sporozoite- and vaccine-induced protection in murine infection models. In BALB/c mice, where SYIPSAEKI is efficiently presented in the context of the major histocompatibility complex class I (MHC-I) molecule H-2-Kd, we established that epitope-specific CD8+ T cell responses contribute to parasite killing following sporozoite immunization. Yet, sterile protection was achieved in the absence of this epitope, substantiating the concept that other antigens can be sufficient for parasite-induced protective immunity. Furthermore, we demonstrated that SYIPSAEKI-specific CD8+ T cell responses elicited by viral-vectored CSP-expressing vaccines effectively targeted parasites in hepatocytes. The resulting sterile protection strictly relied on the expression of SYIPSAEKI. In C57BL/6 mice, which are unable to present the immunodominant epitope, CSP-based vaccines did not confer complete protection, despite the induction of high levels of CSP-specific antibodies. These findings underscore the significance of CSP in protection against malaria pre-erythrocytic stages and demonstrate that a significant proportion of the protection against the parasite is mediated by CD8+ T cells specific for the immunodominant CSP-derived epitope.
Collapse
|
16
|
Live Visualization of Hemagglutinin Dynamics During Infection by Using a Novel Reporter Influenza A Virus. Viruses 2020; 12:v12060687. [PMID: 32604762 PMCID: PMC7354568 DOI: 10.3390/v12060687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Live visualization of influenza A virus (IAV) structural proteins during viral infection in cells is highly sought objective to study different aspects of the viral replication cycle. To achieve this, we engineered an IAV to express a Tetra Cysteine tag (TC tag) from hemagglutinin (HA), which allows intracellular labeling of the engineered HA protein with biarsenic dyes and subsequent fluorescence detection. Using such constructs, we rescued a recombinant IAV with TC tag inserted in HA, in A/Puerto Rico/8/1934(H1N1) background (HA-TC). This recombinant HA-TC tag reporter IAV was replication-competent; however, as compared to wild type PR8 IAV, it was attenuated in multicycle replication. We confirmed expression of TC tag and biarsenical labeling of HA by immunofluorescence assay in cells infected with an HA-TC tag reporter IAV. Further, we used this reporter virus to visualize HA expression and translocation in IAV infected cells by live confocal imaging. We also tested the utility of the HA-TC IAV in testing chemical inhibitors of the HA translocation. Overall, HA-TC IAV is a versatile tool that will be useful for studying viral life cycle events, virus-host interactions, and anti-viral testing.
Collapse
|
17
|
Immunization with DNA prime-subunit protein boost strategy based on influenza H9N2 virus conserved matrix protein M1 and its epitope screening. Sci Rep 2020; 10:4144. [PMID: 32139720 PMCID: PMC7057951 DOI: 10.1038/s41598-020-60783-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/17/2020] [Indexed: 12/23/2022] Open
Abstract
Developing an effective universal influenza vaccine against influenza virus with highly conserved antigenic epitopes could induce a broad-spectrum immune response to prevent infection. The soluble protein M1 that can induce the M1 specific immune response was first confirmed in our previous study. In this study, we characterized the immune response induced by DNA prime-subunit protein boost strategy based on the relatively conserved matrix protein 1 (M1) in the BALB/c mouse model, and evaluated its protection ability against a lethal challenge of homologous H9N2 avian influenza virus (A/Chicken/Jiangsu/11/2002). The results showed that 100 μg DNA prime + 100 μg M1 subunit protein boost-strategy significantly increased antibody levels more than vaccination with M1 DNA or M1 subunit protein alone, and induced a more balanced Th1 / Th2 immune response, which not only can provide protection against the homologous virus but also can provide part of the cross-protection against the heterosubtypic PR8 H1N1 strain. In addition, we used an Elispot assay to preliminary screen the T cell epitope in M1 protein, and identified that p22 (M111-25 VLSIIPSGPLKAEIA) epitope was the only immunodominant M1-specific CD4+ T cell epitopes, which could be helpful in understanding the function of influenza virus T cell epitopes.
Collapse
|
18
|
Valdés I, Lazo L, Hermida L, Guillén G, Gil L. Can Complementary Prime-Boost Immunization Strategies Be an Alternative and Promising Vaccine Approach Against Dengue Virus? Front Immunol 2019; 10:1956. [PMID: 31507591 PMCID: PMC6718459 DOI: 10.3389/fimmu.2019.01956] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/02/2019] [Indexed: 12/27/2022] Open
Abstract
Dengue is one of the most important diseases transmitted by mosquitoes. Dengvaxia®, a vaccine registered in several countries, cannot be administered to non-immune individuals and children younger than 9 years old, due to safety reasons. There are two vaccine candidates in phase 3 efficacy trials, but their registration date is completely unknown at this moment. So, the development of new vaccines or vaccine strategies continues to be a priority for the WHO. This work reviews some complementary prime-boost immunization studies against important human pathogens. Additionally, it reviews the results obtained using this regimen of immunization against dengue virus as a potential alternative approach for finding a safe and efficient vaccine. Finally, the main elements associated with this strategy are also discussed. The generation of new strategies of vaccination against dengue virus, must be directed to reduce the risk of increasing viral load through sub-neutralizing antibodies and it must be also directed to induce a polyfunctional T cell response. Complementary prime-boost immunization strategies could emerge as an interesting approach to induce solid immunity or at least to reduce viral load after natural infection, avoiding severe dengue. Subunit vaccine could be safe and attractive antigens for this strategy, especially proteins including B, and T-cells epitopes for inducing humoral and cellular immune responses, which can play an important role controlling the disease.
Collapse
Affiliation(s)
- Iris Valdés
- Vaccine Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Laura Lazo
- Vaccine Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Lisset Hermida
- Vaccine Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gerardo Guillén
- Vaccine Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Lázaro Gil
- Vaccine Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
19
|
Yang H, Yan Z, Zhang Z, Realivazquez A, Ma B, Liu Y. Anti-caries vaccine based on clinical cold-adapted influenza vaccine: A promising alternative for scientific and public-health protection against dental caries. Med Hypotheses 2019; 126:42-45. [PMID: 31010498 DOI: 10.1016/j.mehy.2019.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/21/2019] [Accepted: 03/20/2019] [Indexed: 01/20/2023]
Abstract
Dental caries remains one of the most pervasive infectious disease around the world. Protection against dental caries can be achieved experimentally by eliciting salivary IgA targeting surficial antigens of S. mutans, however, no such a vaccine has been launched for human use yet. Live vectored vaccines hold the greatest feasibility to induce potent and long-lasting immunity in the host. The FDA approved intranasal cold-adapted influenza vaccine has been used in clinical settings for many years. The vaccine can not only induce broad adaptive immune responses especially mucosal immunity, but the member strains can also circumvent existing immunity, presenting promising candidates for live vectored anti-caries vaccine. Moreover, the genetic techniques for modification of cold-adapted influenza viruses are well developed and widely applicable. Thus, we hypothesize that effective anti-caries vaccine can be developed with the backbone of cold-adapted influenza viruses by inserting specific antigenic identifier sequences of S. mutans into the viral genome, which is anticipated to protect against dental caries in humans with easy inoculation. The immune efficacies of recombinant cold-adapted influenza viruses expressing exogenous antigens have been verified by in vivo experiments for multiple infectious diseases, giving us great confidence to validate the safety properties and protection effect with this chimeric vaccine in animals or even humans. Existing data suggests that the live anti-caries vaccine may help improve public oral health by controlling the caries disease itself.
Collapse
Affiliation(s)
- Huixiao Yang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, PR China
| | - Zhonghai Yan
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Zijian Zhang
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Adilene Realivazquez
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Binger Ma
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, PR China
| | - Yi Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610065, PR China.
| |
Collapse
|
20
|
Gerlach T, Elbahesh H, Saletti G, Rimmelzwaan GF. Recombinant influenza A viruses as vaccine vectors. Expert Rev Vaccines 2019; 18:379-392. [PMID: 30777467 DOI: 10.1080/14760584.2019.1582338] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Various viruses, including poxviruses, adenoviruses and vesicular stomatitis virus, have been considered as vaccine vectors for the delivery of antigens of interest in the development of vaccines against newly emerging pathogens. AREAS COVERED Here, we review results that have been obtained with influenza A viruses (IAV) as vaccine vectors. With the advent of reverse genetics technology, IAV-based recombinant vaccine candidates have been constructed that induce protective immunity to a variety of different pathogens of interest, including West Nile virus, Plasmodium falciparum and respiratory syncytial virus. The various cloning strategies to produce effective and attenuated, safe to use IAV-based viral vectors are discussed. EXPERT COMMENTARY It was concluded that IAV-based vector system has several advantages and holds promise for further development.
Collapse
Affiliation(s)
- Thomas Gerlach
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Husni Elbahesh
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Giulietta Saletti
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Guus F Rimmelzwaan
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| |
Collapse
|
21
|
Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199-1211. [PMID: 30333613 DOI: 10.1038/s41590-018-0228-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023]
Abstract
Development of a malaria vaccine remains a critical priority to decrease clinical disease and mortality and facilitate eradication. Accordingly, RTS,S, a protein-subunit vaccine, has completed phase III clinical trials and confers ~30% protection against clinical infection over 4 years. Whole-attenuated-sporozoite and viral-subunit vaccines induce between 20% and 100% protection against controlled human malaria infection, but there is limited published evidence to date for durable, high-level efficacy (>50%) against natural exposure. Importantly, fundamental scientific advances related to the potency, durability, breadth and location of immune responses will be required for improving vaccine efficacy with these and other vaccine approaches. In this Review, we focus on the current understanding of immunological mechanisms of protection from animal models and human vaccine studies, and on how these data should inform the development of next-generation vaccines. Furthermore, we introduce the concept of using passive immunization with monoclonal antibodies as a new approach to prevent and eliminate malaria.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
22
|
Gola A, Silman D, Walters AA, Sridhar S, Uderhardt S, Salman AM, Halbroth BR, Bellamy D, Bowyer G, Powlson J, Baker M, Venkatraman N, Poulton I, Berrie E, Roberts R, Lawrie AM, Angus B, Khan SM, Janse CJ, Ewer KJ, Germain RN, Spencer AJ, Hill AVS. Prime and target immunization protects against liver-stage malaria in mice. Sci Transl Med 2018; 10:10/460/eaap9128. [DOI: 10.1126/scitranslmed.aap9128] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/08/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022]
Abstract
Despite recent advances in treatment and vector control, malaria is still a leading cause of death, emphasizing the need for an effective vaccine. The malaria life cycle can be subdivided into three stages: the invasion and growth within liver hepatocytes (pre-erythrocytic stage), the blood stage (erythrocytic stage), and, finally, the sexual stage (occurring within the mosquito vector). Antigen (Ag)-specific CD8+ T cells are effectively induced by heterologous prime-boost viral vector immunization and known to correlate with liver-stage protection. However, liver-stage malaria vaccines have struggled to generate and maintain the high numbers of Plasmodium-specific circulating T cells necessary to confer sterile protection. We describe an alternative “prime and target” vaccination strategy aimed specifically at inducing high numbers of tissue-resident memory T cells present in the liver at the time of hepatic infection. This approach bypasses the need for very high numbers of circulating T cells and markedly increases the efficacy of subunit immunization against liver-stage malaria with clinically relevant Ags and clinically tested viral vectors in murine challenge models. Translation to clinical use has begun, with encouraging results from a pilot safety and feasibility trial of intravenous chimpanzee adenovirus vaccination in humans. This work highlights the value of a prime-target approach for immunization against malaria and suggests that this strategy may represent a more general approach for prophylaxis or immunotherapy of other liver infections and diseases.
Collapse
|
23
|
Shiratsuchi T, Rai U, Kaneko I, Zhang M, Iwanaga S, Yuda M, Tsuji M. A potent malaria vaccine based on adenovirus with dual modifications at Hexon and pVII. Vaccine 2017; 35:6990-7000. [PMID: 29089194 DOI: 10.1016/j.vaccine.2017.10.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/23/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
Abstract
Adenovirus (Ad) is thought to be one of the most promising platforms for a malaria vaccine targeted against its liver stages, because of its ability to induce a strong T-cell response against a transgene. However, a further improvement of this platform is needed in order to elicit another arm of the immunity, i.e. humoral response, against malaria. In order to augment immunogenicity and protective efficacy of Ad-based malaria vaccine, we inserted B-cell, as well as CD4+ T-cell, epitopes of Plasmodium falciparum circumsporozoite protein (PfCSP) into the capsid protein, Hexon, and the core protein, VII (pVII), of Ad, respectively, in addition to the PfCSP transgene. Insertion of PfCSP-derived B cell epitope to Hexon significantly enhanced the epitope-specific antibody response compared to AdPfCSP, an Ad vaccine expressing only PfCSP transgene. PfCSP-derived CD4+ T-cell epitope insertion into pVII augmented not only PfCSP-specific CD4+ T-cell response but also anti-PfCSP antibody response. Finally, mice immunized with AdPfCSP having both Hexon and pVII modifications were more protected than AdPfCSP or Hexon-modified AdPfCSP against challenge with transgenic rodent malaria parasites expressing the PfCSP. Overall, this study has demonstrated that Hexon and pVII-modified AdPfCSP vaccine is a promising malaria vaccine which induces strong PfCSP-specific humoral, CD4+ T-cell, and CD8+ T-cell responses and protects against infection with transgenic malaria parasites expressing the PfCSP.
Collapse
Affiliation(s)
- Takayuki Shiratsuchi
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA; Otsuka Maryland Medicinal Laboratories, Inc., 9900 Medical Center Drive, Rockville, MD 20850, USA
| | - Urvashi Rai
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA
| | - Izumi Kaneko
- Department of Medical Zoology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Min Zhang
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA; Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Shiroh Iwanaga
- Department of Medical Zoology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; Department of Environmental Parasitology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masao Yuda
- Department of Medical Zoology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Moriya Tsuji
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, 455 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
24
|
Abstract
In 2013 there were an estimated 584,000 deaths and 198 million clinical illnesses due to malaria, the majority in sub-Saharan Africa. Vaccines would be the ideal addition to the existing armamentarium of anti-malaria tools. However, malaria is caused by parasites, and parasites are much more complex in terms of their biology than the viruses and bacteria for which we have vaccines, passing through multiple stages of development in the human host, each stage expressing hundreds of unique antigens. This complexity makes it more difficult to develop a vaccine for parasites than for viruses and bacteria, since an immune response targeting one stage may not offer protection against a later stage, because different antigens are the targets of protective immunity at different stages. Furthermore, depending on the life cycle stage and whether the parasite is extra- or intra-cellular, antibody and/or cellular immune responses provide protection. It is thus not surprising that there is no vaccine on the market for prevention of malaria, or any human parasitic infection. In fact, no vaccine for any disease with this breadth of targets and immune responses exists. In this limited review, we focus on four approaches to malaria vaccines, (1) a recombinant protein with adjuvant vaccine aimed at Plasmodium falciparum (Pf) pre-erythrocytic stages of the parasite cycle (RTS,S/AS01), (2) whole sporozoite vaccines aimed at Pf pre-erythrocytic stages (PfSPZ Vaccine and PfSPZ-CVac), (3) prime boost vaccines that include recombinant DNA, viruses and bacteria, and protein with adjuvant aimed primarily at Pf pre-erythrocytic, but also asexual erythrocytic stages, and (4) recombinant protein with adjuvant vaccines aimed at Pf and Plasmodium vivax sexual erythrocytic and mosquito stages. We recognize that we are not covering all approaches to malaria vaccine development, or most of the critically important work on development of vaccines against P. vivax, the second most important cause of malaria. Progress during the last few years has been significant, and a first generation malaria candidate vaccine, RTS,S/AS01, is under review by the European Medicines Agency (EMA) for its quality, safety and efficacy under article 58, which allows the EMA to give a scientific opinion about products intended exclusively for markets outside of the European Union. However, much work is in progress to optimize malaria vaccines in regard to magnitude and durability of protective efficacy and the financing and practicality of delivery. Thus, we are hopeful that anti-malaria vaccines will soon be important tools in the battle against malaria.
Collapse
|
25
|
Abstract
A safe and effective malaria vaccine is a crucial part of the roadmap to malaria elimination/eradication by the year 2050. Viral-vectored vaccines based on adenoviruses and modified vaccinia virus Ankara (MVA) expressing malaria immunogens are currently being used in heterologous prime-boost regimes in clinical trials for induction of strong antigen-specific T-cell responses and high-titer antibodies. Recombinant MVA is a safe and well-tolerated attenuated vector that has consistently shown significant boosting potential. Advances have been made in large-scale MVA manufacture as high-yield producer cell lines and high-throughput purification processes have recently been developed. This review describes the use of MVA as malaria vaccine vector in both preclinical and clinical studies in the past 5 years.
Collapse
|
26
|
Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine 2015; 33 Suppl 4:D13-23. [PMID: 26324116 DOI: 10.1016/j.vaccine.2015.07.091] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/25/2015] [Accepted: 07/27/2015] [Indexed: 01/14/2023]
Abstract
In 2013 there were an estimated 584,000 deaths and 198 million clinical illnesses due to malaria, the majority in sub-Saharan Africa. Vaccines would be the ideal addition to the existing armamentarium of anti-malaria tools. However, malaria is caused by parasites, and parasites are much more complex in terms of their biology than the viruses and bacteria for which we have vaccines, passing through multiple stages of development in the human host, each stage expressing hundreds of unique antigens. This complexity makes it more difficult to develop a vaccine for parasites than for viruses and bacteria, since an immune response targeting one stage may not offer protection against a later stage, because different antigens are the targets of protective immunity at different stages. Furthermore, depending on the life cycle stage and whether the parasite is extra- or intra-cellular, antibody and/or cellular immune responses provide protection. It is thus not surprising that there is no vaccine on the market for prevention of malaria, or any human parasitic infection. In fact, no vaccine for any disease with this breadth of targets and immune responses exists. In this limited review, we focus on four approaches to malaria vaccines, (1) a recombinant protein with adjuvant vaccine aimed at Plasmodium falciparum (Pf) pre-erythrocytic stages of the parasite cycle (RTS,S/AS01), (2) whole sporozoite vaccines aimed at Pf pre-erythrocytic stages (PfSPZ Vaccine and PfSPZ-CVac), (3) prime boost vaccines that include recombinant DNA, viruses and bacteria, and protein with adjuvant aimed primarily at Pf pre-erythrocytic, but also asexual erythrocytic stages, and (4) recombinant protein with adjuvant vaccines aimed at Pf and Plasmodium vivax sexual erythrocytic and mosquito stages. We recognize that we are not covering all approaches to malaria vaccine development, or most of the critically important work on development of vaccines against P. vivax, the second most important cause of malaria. Progress during the last few years has been significant, and a first generation malaria candidate vaccine, RTS,S/AS01, is under review by the European Medicines Agency (EMA) for its quality, safety and efficacy under article 58, which allows the EMA to give a scientific opinion about products intended exclusively for markets outside of the European Union. However, much work is in progress to optimize malaria vaccines in regard to magnitude and durability of protective efficacy and the financing and practicality of delivery. Thus, we are hopeful that anti-malaria vaccines will soon be important tools in the battle against malaria.
Collapse
Affiliation(s)
| | | | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
27
|
Kittel C, Wressnigg N, Shurygina AP, Wolschek M, Stukova M, Romanovskaya-Romanko E, Romanova J, Kiselev O, Muster T, Egorov A. A genetically adjuvanted influenza B virus vector increases immunogenicity and protective efficacy in mice. Arch Virol 2015. [PMID: 26215439 DOI: 10.1007/s00705-015-2525-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The existence of multiple antigenically distinct types and subtypes of influenza viruses allows the construction of a multivalent vector system for the mucosal delivery of foreign sequences. Influenza A viruses have been exploited successfully for the expression of extraneous antigens as well as immunostimulatory molecules. In this study, we describe the development of an influenza B virus vector whose functional part of the interferon antagonist NS1 was replaced by human interleukin 2 (IL2) as a genetic adjuvant. We demonstrate that IL2 expressed by this viral vector displays immune adjuvant activity in immunized mice. Animals vaccinated with the IL2 viral vector showed an increased hemagglutination inhibition antibody response and higher protective efficacy after challenge with a wild-type influenza B virus when compared to mice vaccinated with a control virus. Our results demonstrate that it is feasible to construct influenza B vaccine strains expressing immune-potentiating foreign sequences from the NS genomic segment. Based on these data, it is now hypothetically possible to create a trivalent (or quadrivalent) live attenuated influenza vaccine in which each component expresses a selected genetic adjuvant with tailored expression levels.
Collapse
Affiliation(s)
- Christian Kittel
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria.
| | - Nina Wressnigg
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria
| | - Anna Polina Shurygina
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria
- Influenza Research Institute, Russian Academy of Medical Sciences, Prof. Popov Str. 15/17, St. Petersburg, 197376, Russia
| | - Markus Wolschek
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria
| | - Marina Stukova
- Influenza Research Institute, Russian Academy of Medical Sciences, Prof. Popov Str. 15/17, St. Petersburg, 197376, Russia
| | | | - Julia Romanova
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria
| | - Oleg Kiselev
- Influenza Research Institute, Russian Academy of Medical Sciences, Prof. Popov Str. 15/17, St. Petersburg, 197376, Russia
| | - Thomas Muster
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria
| | - Andrej Egorov
- Avir Green Hills Biotechnology AG, Forsthausgasse 11, 1200, Vienna, Austria
| |
Collapse
|
28
|
García-Arriaza J, Esteban M. Enhancing poxvirus vectors vaccine immunogenicity. Hum Vaccin Immunother 2015; 10:2235-44. [PMID: 25424927 DOI: 10.4161/hv.28974] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Attenuated recombinant poxvirus vectors expressing heterologous antigens from pathogens are currently at various stages in clinical trials with the aim to establish their efficacy. This is because these vectors have shown excellent safety profiles, significant immunogenicity against foreign expressed antigens and are able to induce protective immune responses. In view of the limited efficacy triggered by some poxvirus strains used in clinical trials (i.e, ALVAC in the RV144 phase III clinical trial for HIV), and of the restrictive replication capacity of the highly attenuated vectors like MVA and NYVAC, there is a consensus that further improvements of these vectors should be pursuit. In this review we considered several strategies that are currently being implemented, as well as new approaches, to improve the immunogenicity of the poxvirus vectors. This includes heterologous prime/boost protocols, use of co-stimulatory molecules, deletion of viral immunomodulatory genes still present in the poxvirus genome, enhancing virus promoter strength, enhancing vector replication capacity, optimizing expression of foreign heterologous sequences, and the combined use of adjuvants. An optimized poxvirus vector triggering long-lasting immunity with a high protective efficacy against a selective disease should be sought.
Collapse
Affiliation(s)
- Juan García-Arriaza
- a Department of Molecular and Cellular Biology; Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CSIC); Madrid, Spain
| | | |
Collapse
|
29
|
Noe AR, Espinosa D, Li X, Coelho-dos-Reis JGA, Funakoshi R, Giardina S, Jin H, Retallack DM, Haverstock R, Allen JR, Vedvick TS, Fox CB, Reed SG, Ayala R, Roberts B, Winram SB, Sacci J, Tsuji M, Zavala F, Gutierrez GM. A full-length Plasmodium falciparum recombinant circumsporozoite protein expressed by Pseudomonas fluorescens platform as a malaria vaccine candidate. PLoS One 2014; 9:e107764. [PMID: 25247295 PMCID: PMC4172688 DOI: 10.1371/journal.pone.0107764] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/14/2014] [Indexed: 11/19/2022] Open
Abstract
The circumsporozoite protein (CSP) of Plasmodium falciparum is a major surface protein, which forms a dense coat on the sporozoite's surface. Preclinical research on CSP and clinical evaluation of a CSP fragment-based RTS, S/AS01 vaccine have demonstrated a modest degree of protection against P. falciparum, mediated in part by humoral immunity and in part by cell-mediated immunity. Given the partial protective efficacy of the RTS, S/AS01 vaccine in a recent Phase 3 trial, further improvement of CSP-based vaccines is crucial. In this report, we describe the preclinical development of a full-length, recombinant CSP (rCSP)-based vaccine candidate against P. falciparum malaria suitable for current Good Manufacturing Practice (cGMP) production. Utilizing a novel high-throughput Pseudomonas fluorescens expression platform, we demonstrated greater efficacy of full-length rCSP as compared to N-terminally truncated versions, rapidly down-selected a promising lead vaccine candidate, and developed a high-yield purification process to express immunologically active, intact antigen for clinical trial material production. The rCSP, when formulated with various adjuvants, induced antigen-specific antibody responses as measured by enzyme-linked immunosorbent assay (ELISA) and immunofluorescence assay (IFA), as well as CD4+ T-cell responses as determined by ELISpot. The adjuvanted rCSP vaccine conferred protection in mice when challenged with transgenic P. berghei sporozoites containing the P. falciparum repeat region of CSP. Furthermore, heterologous prime/boost regimens with adjuvanted rCSP and an adenovirus type 35-vectored CSP (Ad35CS) showed modest improvements in eliciting CSP-specific T-cell responses and anti-malarial protection, depending on the order of vaccine delivery. Collectively, these data support the importance of further clinical development of adjuvanted rCSP, either as a stand-alone product or as one of the components in a heterologous prime/boost strategy, ultimately acting as an effective vaccine candidate for the mitigation of P. falciparum-induced malaria.
Collapse
Affiliation(s)
- Amy R. Noe
- Leidos Inc., Frederick, Maryland, United States of America
| | - Diego Espinosa
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Xiangming Li
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Jordana G. A. Coelho-dos-Reis
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Ryota Funakoshi
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Steve Giardina
- Leidos Inc., Frederick, Maryland, United States of America
| | - Hongfan Jin
- Pfenex Inc., San Diego, California, United States of America
| | | | - Ryan Haverstock
- Pfenex Inc., San Diego, California, United States of America
| | | | - Thomas S. Vedvick
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Christopher B. Fox
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Ramses Ayala
- Leidos Inc., Frederick, Maryland, United States of America
| | - Brian Roberts
- Leidos Inc., Frederick, Maryland, United States of America
| | | | - John Sacci
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Moriya Tsuji
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Fidel Zavala
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | | |
Collapse
|
30
|
Ondondo BO. The influence of delivery vectors on HIV vaccine efficacy. Front Microbiol 2014; 5:439. [PMID: 25202303 PMCID: PMC4141443 DOI: 10.3389/fmicb.2014.00439] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/03/2014] [Indexed: 12/31/2022] Open
Abstract
Development of an effective HIV/AIDS vaccine remains a big challenge, largely due to the enormous HIV diversity which propels immune escape. Thus novel vaccine strategies are targeting multiple variants of conserved antibody and T cell epitopic regions which would incur a huge fitness cost to the virus in the event of mutational escape. Besides immunogen design, the delivery modality is critical for vaccine potency and efficacy, and should be carefully selected in order to not only maximize transgene expression, but to also enhance the immuno-stimulatory potential to activate innate and adaptive immune systems. To date, five HIV vaccine candidates have been evaluated for efficacy and protection from acquisition was only achieved in a small proportion of vaccinees in the RV144 study which used a canarypox vector for delivery. Conversely, in the STEP study (HVTN 502) where human adenovirus serotype 5 (Ad5) was used, strong immune responses were induced but vaccination was more associated with increased risk of HIV acquisition than protection in vaccinees with pre-existing Ad5 immunity. The possibility that pre-existing immunity to a highly promising delivery vector may alter the natural course of HIV to increase acquisition risk is quite worrisome and a huge setback for HIV vaccine development. Thus, HIV vaccine development efforts are now geared toward delivery platforms which attain superior immunogenicity while concurrently limiting potential catastrophic effects likely to arise from pre-existing immunity or vector-related immuno-modulation. However, it still remains unclear whether it is poor immunogenicity of HIV antigens or substandard immunological potency of the safer delivery vectors that has limited the success of HIV vaccines. This article discusses some of the promising delivery vectors to be harnessed for improved HIV vaccine efficacy.
Collapse
Affiliation(s)
- Beatrice O Ondondo
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford Oxford, UK
| |
Collapse
|
31
|
Abstract
Reverse genetics systems allow artificial generation of non-segmented and segmented negative-sense RNA viruses, like influenza viruses, entirely from cloned cDNA. Since the introduction of reverse genetics systems over a decade ago, the ability to generate ‘designer’ influenza viruses in the laboratory has advanced both basic and applied research, providing a powerful tool to investigate and characterise host–pathogen interactions and advance the development of novel therapeutic strategies. The list of applications for reverse genetics has expanded vastly in recent years. In this review, we discuss the development and implications of this technique, including the recent controversy surrounding the generation of a transmissible H5N1 influenza virus. We will focus on research involving the identification of viral protein function, development of live-attenuated influenza virus vaccines, host–pathogen interactions, immunity and the generation of recombinant influenza virus vaccine vectors for the prevention and treatment of infectious diseases and cancer.
Collapse
|
32
|
CD8+ T cells eliminate liver-stage Plasmodium berghei parasites without detectable bystander effect. Infect Immun 2014; 82:1460-4. [PMID: 24421043 DOI: 10.1128/iai.01500-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immunization with attenuated Plasmodium sporozoites or viral vectored vaccines can induce protective CD8(+) T cells that can find and eliminate liver-stage malaria parasites. A key question is whether CD8(+) T cells must recognize and eliminate each parasite in the liver or whether bystander killing can occur. To test this, we transferred antigen-specific effector CD8(+) T cells to mice that were then coinfected with two Plasmodium berghei strains, only one of which could be recognized directly by the transferred T cells. We found that the noncognate parasites developed normally in these mice, demonstrating that bystander killing of parasites does not occur during the CD8(+) T cell response to malaria parasites. Rather, elimination of infected parasites is likely mediated by direct recognition of infected hepatocytes by antigen-specific CD8(+) T cells.
Collapse
|
33
|
Abstract
The liver is the largest organ in the body and is generally regarded by nonimmunologists as having little or no lymphoid function. However, such is far from accurate. This review highlights the importance of the liver as a lymphoid organ. Firstly, we discuss experimental data surrounding the role of liver as a lymphoid organ. The liver facilitates tolerance rather than immunoreactivity, which protects the host from antigenic overload of dietary components and drugs derived from the gut and it is instrumental to fetal immune tolerance. Loss of liver tolerance leads to autoaggressive phenomena, which if not controlled by regulatory lymphoid populations, may lead to the induction of autoimmune liver diseases. Liver-related lymphoid subpopulations also act as critical antigen-presenting cells. The study of the immunological properties of liver and delineation of the microenvironment of the intrahepatic milieu in normal and diseased livers provides a platform to understand the hierarchy of a series of detrimental events that lead to immune-mediated destruction of the liver and the rejection of liver allografts. The majority of emphasis within this review will be on the normal mononuclear cell composition of the liver. However, within this context, we will discuss selected, but not all, immune-mediated liver disease and attempt to place these data in the context of human autoimmunity.
Collapse
Affiliation(s)
- Dimitrios P Bogdanos
- Institute of Liver Studies, Transplantation Immunology and Mucosal Biology, King's College London School of Medicine at King's College Hospital, London, UK
| | | | | |
Collapse
|
34
|
Hafalla JCR, Bauza K, Friesen J, Gonzalez-Aseguinolaza G, Hill AVS, Matuschewski K. Identification of targets of CD8⁺ T cell responses to malaria liver stages by genome-wide epitope profiling. PLoS Pathog 2013; 9:e1003303. [PMID: 23675294 PMCID: PMC3649980 DOI: 10.1371/journal.ppat.1003303] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/27/2013] [Indexed: 12/25/2022] Open
Abstract
CD8⁺ T cells mediate immunity against Plasmodium liver stages. However, the paucity of parasite-specific epitopes of CD8⁺ T cells has limited our current understanding of the mechanisms influencing the generation, maintenance and efficiency of these responses. To identify antigenic epitopes in a stringent murine malaria immunisation model, we performed a systematic profiling of H(2b)-restricted peptides predicted from genome-wide analysis. We describe the identification of Plasmodium berghei (Pb) sporozoite-specific gene 20 (S20)- and thrombospondin-related adhesive protein (TRAP)-derived peptides, termed PbS20₃₁₈ and PbTRAP₁₃₀ respectively, as targets of CD8⁺ T cells from C57BL/6 mice vaccinated by whole parasite strategies known to protect against sporozoite challenge. While both PbS20₃₁₈ and PbTRAP₁₃₀ elicit effector and effector memory phenotypes in both the spleens and livers of immunised mice, only PbTRAP₁₃₀-specific CD8⁺ T cells exhibit in vivo cytotoxicity. Moreover, PbTRAP₁₃₀-specific, but not PbS20₃₁₈-specific, CD8⁺ T cells significantly contribute to inhibition of parasite development. Prime/boost vaccination with PbTRAP demonstrates CD8⁺ T cell-dependent efficacy against sporozoite challenge. We conclude that PbTRAP is an immunodominant antigen during liver-stage infection. Together, our results underscore the presence of CD8⁺ T cells with divergent potencies against distinct Plasmodium liver-stage epitopes. Our identification of antigen-specific CD8⁺ T cells will allow interrogation of the development of immune responses against malaria liver stages.
Collapse
Affiliation(s)
- Julius Clemence R. Hafalla
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail: (JCRH); (KM)
| | - Karolis Bauza
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Johannes Friesen
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Gloria Gonzalez-Aseguinolaza
- Department of Gene Therapy and Hepatology, Center for Investigation in Applied Medicine (CIMA), University of Navarra, Pamplona, Spain
| | - Adrian V. S. Hill
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Institute of Biology, Humboldt University, Berlin, Germany
- * E-mail: (JCRH); (KM)
| |
Collapse
|
35
|
Soares IS, Françoso KS, Jampaulo VO, Rodrigues MM. CD8(+) T-cell-mediated immunity against malaria: a novel heterologous prime-boost strategy. Expert Rev Vaccines 2013; 11:1039-41. [PMID: 23151162 DOI: 10.1586/erv.12.82] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recently, a vaccine against malaria was successfully tested in a human Phase III trial. The efficacy of this vaccine formulation, based on the Plasmodium falciparum circumsporozoite protein, was approximately 50% and correlated with the presence of antibodies specific to the infective stages of the malaria parasites. Different strategies are being pursued to improve vaccine efficacy levels. One such strategy is the induction of specific cytotoxic T cells that can destroy the intracellular hepatocyte stages of the malaria parasite. In this study, a novel vaccination protocol was developed to elicit strong immune responses mediated by CD8(+) cytotoxic cells specific to the circumsporozoite protein. As proof-of-concept, the authors used the rodent malaria Plasmodium yoelii parasite. The vaccination strategy consisted of a heterologous prime-boost vaccination regimen involving porcine parvovirus-like particles for priming and the modified vaccinia virus Ankara for the booster immunization, both of which expressed the immunodominant CD8 epitope of the P. yoelii circumsporozoite protein. Results from this experimental model were extremely meaningful. This vaccination strategy led to a significant T-cell immune response mediated by CD8(+) multifunctional T effector and effector-memory cells. However, most importantly for the malaria vaccine development was the fact that following a sporozoite challenge, immunized mice eliminated more than 97% of the malaria parasites during the hepatocyte stages. These results confirm and extend a vast body of knowledge showing that a heterologous prime-boost vaccination strategy can elicit strong CD8(+) T-cell-mediated protective immunity and may increase the efficacy of malaria vaccines.
Collapse
Affiliation(s)
- Irene S Soares
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, São Paulo, 05508-900, Brazil.
| | | | | | | |
Collapse
|
36
|
Attenuated and replication-competent vaccinia virus strains M65 and M101 with distinct biology and immunogenicity as potential vaccine candidates against pathogens. J Virol 2013; 87:6955-74. [PMID: 23596295 DOI: 10.1128/jvi.03013-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Replication-competent poxvirus vectors with an attenuation phenotype and with a high immunogenic capacity of the foreign expressed antigen are being pursued as novel vaccine vectors against different pathogens. In this investigation, we have examined the replication and immunogenic characteristics of two vaccinia virus (VACV) mutants, M65 and M101. These mutants were generated after 65 and 101 serial passages of persistently infected Friend erythroleukemia (FEL) cells. In cultured cells of different origins, the mutants are replication competent and have growth kinetics similar to or slightly reduced in comparison with those of the parental Western Reserve (WR) virus strain. In normal and immune-suppressed infected mice, the mutants showed different levels of attenuation and pathogenicity in comparison with WR and modified vaccinia Ankara (MVA) strains. Wide genome analysis after deep sequencing revealed selected genomic deletions and mutations in a number of viral open reading frames (ORFs). Mice immunized in a DNA prime/mutant boost regimen with viral vectors expressing the LACK (Leishmania homologue for receptors of activated C kinase) antigen of Leishmania infantum showed protection or a delay in the onset of cutaneous leishmaniasis. Protection was similar to that triggered by MVA-LACK. In immunized mice, both polyfunctional CD4(+) and CD8(+) T cells with an effector memory phenotype were activated by the two mutants, but the DNA-LACK/M65-LACK protocol preferentially induced CD4(+) whereas DNA-LACK/M101-LACK preferentially induced CD8(+) T cell responses. Altogether, our findings showed the adaptive changes of the WR genome during long-term virus-host cell interaction and how the replication competency of M65 and M101 mutants confers distinct biological properties and immunogenicity in mice compared to those of the MVA strain. These mutants could have applicability for understanding VACV biology and as potential vaccine vectors against pathogens and tumors.
Collapse
|
37
|
Ewer KJ, O’Hara GA, Duncan CJA, Collins KA, Sheehy SH, Reyes-Sandoval A, Goodman AL, Edwards NJ, Elias SC, Halstead FD, Longley RJ, Rowland R, Poulton ID, Draper SJ, Blagborough AM, Berrie E, Moyle S, Williams N, Siani L, Folgori A, Colloca S, Sinden RE, Lawrie AM, Cortese R, Gilbert SC, Nicosia A, Hill AVS. Protective CD8+ T-cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation. Nat Commun 2013; 4:2836. [PMID: 24284865 PMCID: PMC3868203 DOI: 10.1038/ncomms3836] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 10/29/2013] [Indexed: 02/01/2023] Open
Abstract
Induction of antigen-specific CD8(+) T cells offers the prospect of immunization against many infectious diseases, but no subunit vaccine has induced CD8(+) T cells that correlate with efficacy in humans. Here we demonstrate that a replication-deficient chimpanzee adenovirus vector followed by a modified vaccinia virus Ankara booster induces exceptionally high frequency T-cell responses (median >2400 SFC/10(6) peripheral blood mononuclear cells) to the liver-stage Plasmodium falciparum malaria antigen ME-TRAP. It induces sterile protective efficacy against heterologous strain sporozoites in three vaccinees (3/14, 21%), and delays time to patency through substantial reduction of liver-stage parasite burden in five more (5/14, 36%), P=0.008 compared with controls. The frequency of monofunctional interferon-γ-producing CD8(+) T cells, but not antibodies, correlates with sterile protection and delay in time to patency (P(corrected)=0.005). Vaccine-induced CD8(+) T cells provide protection against human malaria, suggesting that a major limitation of previous vaccination approaches has been the insufficient magnitude of induced T cells.
Collapse
Affiliation(s)
- Katie J. Ewer
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
- These authors contributed equally to this work
| | - Geraldine A. O’Hara
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- These authors contributed equally to this work
| | - Christopher J. A. Duncan
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- These authors contributed equally to this work
| | - Katharine A. Collins
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Susanne H. Sheehy
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Anna L. Goodman
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Nick J. Edwards
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Sean C. Elias
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Fenella D. Halstead
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Rhea J. Longley
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Rosalind Rowland
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Ian D. Poulton
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Simon J. Draper
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | | | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford OX3 7JT, UK
| | - Sarah Moyle
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford OX3 7JT, UK
| | - Nicola Williams
- Centre for Statistics in Medicine, Linton Road, Oxford OX2 6UD, UK
| | | | | | | | - Robert E. Sinden
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
- Division of Cell and Molecular Biology, Imperial College London, London SW7 2AZ, UK
| | - Alison M. Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | | | - Sarah C. Gilbert
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Alfredo Nicosia
- Okairos, viale Citta’ d’Europa 279, Rome 00144, Italy
- CEINGE, via Gaetano Salvatore 486, Naples 80145, Italy
- Department of Biochemistry and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
| | - Adrian V. S. Hill
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| |
Collapse
|
38
|
Vasconcelos JR, Dominguez MR, Araújo AF, Ersching J, Tararam CA, Bruna-Romero O, Rodrigues MM. Relevance of long-lived CD8(+) T effector memory cells for protective immunity elicited by heterologous prime-boost vaccination. Front Immunol 2012; 3:358. [PMID: 23264773 PMCID: PMC3525016 DOI: 10.3389/fimmu.2012.00358] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/10/2012] [Indexed: 11/13/2022] Open
Abstract
Owing to the importance of major histocompatibility complex class Ia-restricted CD8(+) T cells for host survival following viral, bacterial, fungal, or parasitic infection, it has become largely accepted that these cells should be considered in the design of a new generation of vaccines. For the past 20 years, solid evidence has been provided that the heterologous prime-boost regimen achieves the best results in terms of induction of long-lived protective CD8(+) T cells against a variety of experimental infections. Although this regimen has often been used experimentally, as is the case for many vaccines, the mechanism behind the efficacy of this vaccination regimen is still largely unknown. The main purpose of this review is to examine the characteristics of the protective CD8(+) T cells generated by this vaccination regimen. Part of its efficacy certainly relies on the generation and maintenance of large numbers of specific lymphocytes. Other specific characteristics may also be important, and studies on this direction have only recently been initiated. So far, the characterization of these protective, long-lived T cell populations suggests that there is a high frequency of polyfunctional T cells; these cells cover a large breadth and display a T effector memory (TEM) phenotype. These TEM cells are capable of proliferating after an infectious challenge and are highly refractory to apoptosis due to a control of the expression of pro-apoptotic receptors such as CD95. Also, they do not undergo significant long-term immunological erosion. Understanding the mechanisms that control the generation and maintenance of the protective activity of these long-lived TEM cells will certainly provide important insights into the physiology of CD8(+) T cells and pave the way for the design of new or improved vaccines.
Collapse
Affiliation(s)
- José R Vasconcelos
- Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo - Escola Paulista de Medicina São Paulo, São Paulo, Brazil ; Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
39
|
Ono T, Yamaguchi Y, Oguma T, Takayama E, Takashima Y, Tadakuma T, Miyahira Y. Actively induced antigen-specific CD8+ T cells by epitope-bearing parasite pre-infection but not prime/boost virus vector vaccination could ameliorate the course of Plasmodium yoelii blood-stage infection. Vaccine 2012; 30:6270-8. [PMID: 22902783 DOI: 10.1016/j.vaccine.2012.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/31/2012] [Accepted: 08/04/2012] [Indexed: 12/16/2022]
Abstract
The lack of MHC molecules on red blood cells (RBCs) has led to questions regarding the immunological function of CD8(+) T cells against malarial blood-stage (MBS). However, several recent reports contradicting with this concept have suggested that they play an important role in the course of MBS infection. The present study generated genetically engineered murine malaria, Plasmodium yoelii, which expresses a well-defined Trypanosoma cruzi-derived, H-2K(b)-restricted CD8(+) T cell epitope, ANYNFTLV. Prime/boost vaccination by the use of recombinant adenovirus and recombinant modified vaccinia virus Ankara (MVA), which induced an enhanced number of ANYNFTLV-specific CD8(+) T cells, failed to prevent a pathological outcome to occur upon ANYNFTLV-expressing murine MBS infection. This outcome did not change even with the combination of passive transfer of an appreciable number of in vitro-expanded ANYNFTLV-specific CD8(+) T cells. In contrast, the pre-infection of mice with T. cruzi, which intrinsically bears the same CD8(+) T cell epitope significantly improved the survival of ANYNFTLV-expressing malaria-infected mice but not that of control malaria-infected ones. This protective effect was abrogated by the use of a CD8(+) T cell-depleting monoclonal antibody. Although the protective effect was observed only in certain situations, the actively induced antigen-specific CD8(+) T cells could ameliorate the pathologies caused by the MBS. This is the first study to implicate that the active induction of antigen-specific CD8(+) T cells should be included in the development of a vaccine against MBS.
Collapse
Affiliation(s)
- Takeshi Ono
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa City, Saitama 359-8513, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Vijayan A, Gómez CE, Espinosa DA, Goodman AG, Sanchez-Sampedro L, Sorzano COS, Zavala F, Esteban M. Adjuvant-like effect of vaccinia virus 14K protein: a case study with malaria vaccine based on the circumsporozoite protein. THE JOURNAL OF IMMUNOLOGY 2012; 188:6407-17. [PMID: 22615208 DOI: 10.4049/jimmunol.1102492] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Development of subunit vaccines for malaria that elicit a strong, long-term memory response is an intensive area of research, with the focus on improving the immunogenicity of a circumsporozoite (CS) protein-based vaccine. In this study, we found that a chimeric protein, formed by fusing vaccinia virus protein 14K (A27) to the CS of Plasmodium yoelii, induces strong effector memory CD8(+) T cell responses in addition to high-affinity Abs when used as a priming agent in the absence of any adjuvant, followed by an attenuated vaccinia virus boost expressing CS in murine models. Moreover, priming with the chimeric protein improved the magnitude and polyfunctionality of cytokine-secreting CD8(+) T cells. This fusion protein formed oligomers/aggregates that led to activation of STAT-1 and IFN regulatory factor-3 in human macrophages, indicating a type I IFN response, resulting in NO, IL-12, and IL-6 induction. Furthermore, this vaccination regimen inhibited the liver stage development of the parasite, resulting in sterile protection. In summary, we propose a novel approach in designing CS based pre-erythrocytic vaccines against Plasmodium using the adjuvant-like effect of the immunogenic vaccinia virus protein 14K.
Collapse
Affiliation(s)
- Aneesh Vijayan
- Departamento de Biología Celular y Molecular, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Rodríguez D, González-Aseguinolaza G, Rodríguez JR, Vijayan A, Gherardi M, Rueda P, Casal JI, Esteban M. Vaccine efficacy against malaria by the combination of porcine parvovirus-like particles and vaccinia virus vectors expressing CS of Plasmodium. PLoS One 2012; 7:e34445. [PMID: 22529915 PMCID: PMC3328484 DOI: 10.1371/journal.pone.0034445] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 03/02/2012] [Indexed: 11/18/2022] Open
Abstract
With the aim to develop an efficient and cost-effective approach to control malaria, we have generated porcine parvovirus-like particles (PPV-VLPs) carrying the CD8(+) T cell epitope (SYVPSAEQI) of the circumsporozoite (CS) protein from Plasmodium yoelii fused to the PPV VP2 capsid protein (PPV-PYCS), and tested in prime/boost protocols with poxvirus vectors for efficacy in a rodent malaria model. As a proof-of concept, we have characterized the anti-CS CD8(+) T cell response elicited by these hybrid PPV-VLPs in BALB/c mice after immunizations with the protein PPV-PYCS administered alone or in combination with recombinant vaccinia virus (VACV) vectors from the Western Reserve (WR) and modified virus Ankara (MVA) strains expressing the entire P. yoelii CS protein. The results of different immunization protocols showed that the combination of PPV-PYCS prime/poxvirus boost was highly immunogenic, inducing specific CD8+ T cell responses to CS resulting in 95% reduction in liver stage parasites two days following sporozoite challenge. In contrast, neither the administration of PPV-PYCS alone nor the immunization with the vectors given in the order poxvirus/VLPs was as effective. The immune profile induced by VLPs/MVA boost was associated with polyfunctional and effector memory CD8+ T cell responses. These findings highlight the use of recombinant parvovirus PPV-PYCS particles as priming agents and poxvirus vectors, like MVA, as booster to enhance specific CD8+ T cell responses to Plasmodium antigens and to control infection. These observations are relevant in the design of T cell-inducing vaccines against malaria.
Collapse
Affiliation(s)
- Dolores Rodríguez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | - Juan R. Rodríguez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Aneesh Vijayan
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Magdalena Gherardi
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | | | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
42
|
Tse SW, Radtke AJ, Zavala F. Induction and maintenance of protective CD8+ T cells against malaria liver stages: implications for vaccine development. Mem Inst Oswaldo Cruz 2012; 106 Suppl 1:172-8. [PMID: 21881772 DOI: 10.1590/s0074-02762011000900022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/20/2011] [Indexed: 11/22/2022] Open
Abstract
CD8+ T cells against malaria liver stages represent a major protective immune mechanism against infection. Following induction in the peripheral lymph nodes by dendritic cells (DCs), these CD8+ T cells migrate to the liver and eliminate parasite infected hepatocytes. The processing and presentation of sporozoite antigen requires TAP mediated transport of major histocompatibility complex class I epitopes to the endoplasmic reticulum. Importantly, in DCs this process is also dependent on endosome-mediated cross presentation while this mechanism is not required for epitope presentation on hepatocytes. Protective CD8+ T cell responses are strongly dependent on the presence of CD4+ T cells and the capacity of sporozoite antigen to persist for a prolonged period of time. While human trials with subunit vaccines capable of inducing antibodies and CD4+ T cell responses have yielded encouraging results, an effective anti-malaria vaccine will likely require vaccine constructs designed to induce protective CD8+ T cells against malaria liver stages.
Collapse
Affiliation(s)
- Sze-Wah Tse
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
43
|
Schwartz L, Brown GV, Genton B, Moorthy VS. A review of malaria vaccine clinical projects based on the WHO rainbow table. Malar J 2012; 11:11. [PMID: 22230255 PMCID: PMC3286401 DOI: 10.1186/1475-2875-11-11] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/09/2012] [Indexed: 12/14/2022] Open
Abstract
Development and Phase 3 testing of the most advanced malaria vaccine, RTS,S/AS01, indicates that malaria vaccine R&D is moving into a new phase. Field trials of several research malaria vaccines have also confirmed that it is possible to impact the host-parasite relationship through vaccine-induced immune responses to multiple antigenic targets using different platforms. Other approaches have been appropriately tested but turned out to be disappointing after clinical evaluation. As the malaria community considers the potential role of a first-generation malaria vaccine in malaria control efforts, it is an apposite time to carefully document terminated and ongoing malaria vaccine research projects so that lessons learned can be applied to increase the chances of success for second-generation malaria vaccines over the next 10 years. The most comprehensive resource of malaria vaccine projects is a spreadsheet compiled by WHO thanks to the input from funding agencies, sponsors and investigators worldwide. This spreadsheet, available from WHO's website, is known as "the rainbow table". By summarizing the published and some unpublished information available for each project on the rainbow table, the most comprehensive review of malaria vaccine projects to be published in the last several years is provided below.
Collapse
Affiliation(s)
- Lauren Schwartz
- Initiative for Vaccine Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Avenue Appia 20, 1211-CH 27, Geneva, Switzerland
| | | | | | | |
Collapse
|
44
|
A human multi-epitope recombinant vaccinia virus as a universal T cell vaccine candidate against influenza virus. PLoS One 2011; 6:e25938. [PMID: 21998725 PMCID: PMC3187825 DOI: 10.1371/journal.pone.0025938] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 09/14/2011] [Indexed: 12/23/2022] Open
Abstract
There is a need to develop a universal vaccine against influenza virus infection to avoid developing new formulations of a seasonal vaccine each year. Many of the vaccine strategies for a universal vaccine target strain-conserved influenza virus proteins, such as the matrix, polymerase, and nucleoproteins, rather than the surface hemagglutinin and neuraminidase proteins. In addition, non-disease-causing viral vectors are a popular choice as a delivery system for the influenza virus antigens. As a proof-of-concept, we have designed a novel influenza virus immunogen based on the NP backbone containing human T cell epitopes for M1, NS1, NP, PB1 and PA proteins (referred as NPmix) as well as a construct containing the conserved regions of influenza virus neuraminidase (N-terminal) and hemagglutinin (C-terminal) (referred as NA-HA). DNA vectors and vaccinia virus recombinants expressing NPmix (WR-NP) or both NPmix plus NA-HA (WR-flu) in the cytosol were tested in a heterologous DNA-prime/vaccinia virus-boost vaccine regimen in mice. We observed an increase in the number of influenza virus-specific IFNγ-secreting splenocytes, composed of populations marked by CD4(+) and CD8(+) T cells producing IFNγ or TNFα. Upon challenge with influenza virus, the vaccinated mice exhibited decreased viral load in the lungs and a delay in mortality. These findings suggest that DNA prime/poxvirus boost with human multi-epitope recombinant influenza virus proteins is a valid approach for a general T-cell vaccine to protect against influenza virus infection.
Collapse
|
45
|
Almeida APMM, Bruna-Romero O. Synergism/complementarity of recombinant adenoviral vectors and other vaccination platforms during induction of protective immunity against malaria. Mem Inst Oswaldo Cruz 2011; 106 Suppl 1:193-201. [PMID: 21881774 DOI: 10.1590/s0074-02762011000900024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/15/2011] [Indexed: 12/19/2022] Open
Abstract
The lack of immunogenicity of most malaria antigens and the complex immune responses required for achieving protective immunity against this infectious disease have traditionally hampered the development of an efficient human malaria vaccine. The current boom in development of recombinant viral vectors and their use in prime-boost protocols that result in enhanced immune outcomes have increased the number of malaria vaccine candidates that access pre-clinical and clinical trials. In the frontline, adenoviruses and poxviruses seem to be giving the best immunization results in experimental animals and their mutual combination, or their combination with recombinant proteins (formulated in adjuvants and given in sequence or being given as protein/virus admixtures), has been shown to reach unprecedented levels of anti-malaria immunity that predictably will be somehow reproduced in the human setting. However, all this optimism was previously seen in the malaria vaccine development field without many real applicable results to date. We describe here the current state-of-the-art in the field of recombinant adenovirus research for malaria vaccine development, in particular referring to their use in combination with other immunogens in heterologous prime-boost protocols, while trying to simultaneously show our contributions and point of view on this subject.
Collapse
|
46
|
Bouillet LÉM, Dias MO, Dorigo NA, Moura AD, Russell B, Nosten F, Renia L, Braga ÉM, Gazzinelli RT, Rodrigues MM, Soares IS, Bruna-Romero O. Long-term humoral and cellular immune responses elicited by a heterologous Plasmodium vivax apical membrane antigen 1 protein prime/adenovirus boost immunization protocol. Infect Immun 2011; 79:3642-52. [PMID: 21730090 PMCID: PMC3165491 DOI: 10.1128/iai.05048-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 06/10/2011] [Indexed: 12/26/2022] Open
Abstract
Apical membrane antigen 1 (AMA-1) is an invasion-related Plasmodium antigen that is expressed during both intracellular and extracellular asexual stages of the parasite's life cycle, making it an ideal target for induction of humoral and cellular immune responses that can protect against malaria. We show here that when it is administered as a recombinant protein (P) in Montanide ISA720 adjuvant, followed by a recombinant human type 5 adenovirus (Ad), intense and long-lasting Plasmodium vivax AMA-1-specific antibody responses (including both IgG1 and IgG2a), as well as proliferative memory T cell responses, can be detected in immunized mice. Memory T cells displayed both central (CD44(hi) CD62L(hi)) and effector (CD44(hi) CD62L(lo)) phenotypes, with the central memory phenotype prevailing (56% of AMA-1-specific proliferating cells). Considering the main traits of the memory immune responses induced against AMA-1, this particular sequence of immunogens (P followed by Ad), but no others (Ad/Ad, Ad/P, or P/P), displayed an optimal synergistic effect. These results give further support to the need for preclinical studies of P. vivax vaccine candidate AMA-1 administered in prime/boost protocols that include recombinant proteins and adenoviral vectors.
Collapse
MESH Headings
- Adenoviridae
- Adjuvants, Immunologic
- Animals
- Antibodies, Protozoan/biosynthesis
- Antibodies, Protozoan/immunology
- Antigens, Protozoan/immunology
- Cytokines/biosynthesis
- Enzyme-Linked Immunosorbent Assay
- Female
- Hyaluronan Receptors/biosynthesis
- Immunity, Cellular
- Immunity, Humoral
- Immunization
- Immunization, Secondary
- Immunologic Memory
- L-Selectin/biosynthesis
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/immunology
- Malaria, Vivax/immunology
- Malaria, Vivax/prevention & control
- Mannitol/administration & dosage
- Mannitol/analogs & derivatives
- Mannitol/immunology
- Mice
- Mice, Inbred BALB C
- Oleic Acids/administration & dosage
- Oleic Acids/immunology
- Plasmodium vivax/immunology
- Protozoan Proteins/immunology
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/immunology
- T-Lymphocytes/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
| | | | | | | | - Bruce Russell
- Singapore Immunology Network, Biopolis, Agency for Science Technology and Research, Singapore
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mae Sot, Tak, Thailand
- Center for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, United Kingdom
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Laurent Renia
- Singapore Immunology Network, Biopolis, Agency for Science Technology and Research, Singapore
| | | | - Ricardo Tostes Gazzinelli
- Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Brazil
- René Rachou Research Center, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Maurício M. Rodrigues
- Department of Clinical Analyses and Toxicology, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S. Soares
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Oscar Bruna-Romero
- Departments of Microbiology
- René Rachou Research Center, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
47
|
Subdominant/cryptic CD8 T cell epitopes contribute to resistance against experimental infection with a human protozoan parasite. PLoS One 2011; 6:e22011. [PMID: 21779365 PMCID: PMC3136500 DOI: 10.1371/journal.pone.0022011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 06/11/2011] [Indexed: 11/19/2022] Open
Abstract
During adaptive immune response, pathogen-specific CD8+ T cells recognize preferentially a small number of epitopes, a phenomenon known as immunodominance. Its biological implications during natural or vaccine-induced immune responses are still unclear. Earlier, we have shown that during experimental infection, the human intracellular pathogen Trypanosoma cruzi restricts the repertoire of CD8+ T cells generating strong immunodominance. We hypothesized that this phenomenon could be a mechanism used by the parasite to reduce the breath and magnitude of the immune response, favoring parasitism, and thus that artificially broadening the T cell repertoire could favor the host. Here, we confirmed our previous observation by showing that CD8+ T cells of H-2a infected mice recognized a single epitope of an immunodominant antigen of the trans-sialidase super-family. In sharp contrast, CD8+ T cells from mice immunized with recombinant genetic vaccines (plasmid DNA and adenovirus) expressing this same T. cruzi antigen recognized, in addition to the immunodominant epitope, two other subdominant epitopes. This unexpected observation allowed us to test the protective role of the immune response to subdominant epitopes. This was accomplished by genetic vaccination of mice with mutated genes that did not express a functional immunodominant epitope. We found that these mice developed immune responses directed solely to the subdominant/cryptic CD8 T cell epitopes and a significant degree of protective immunity against infection mediated by CD8+ T cells. We concluded that artificially broadening the T cell repertoire contributes to host resistance against infection, a finding that has implications for the host-parasite relationship and vaccine development.
Collapse
|
48
|
Abstract
Malaria is a vector-borne infectious disease caused by unicellular parasites of the genus Plasmodium. These obligate intracellular parasites have the unique capacity to infect and replicate within erythrocytes, which are terminally differentiated host cells that lack antigen presentation pathways. Prior to the cyclic erythrocytic infections that cause the characteristic clinical symptoms of malaria, the parasite undergoes an essential and clinically silent expansion phase in the liver. By infecting privileged host cells, employing programs of complex life stage conversions and expressing varying immunodominant antigens, Plasmodium parasites have evolved mechanisms to downmodulate protective immune responses against ongoing and even future infections. Consequently, anti-malaria immunity develops only gradually over many years of repeated and multiple infections in endemic areas. The identification of immune correlates of protection among the abundant non-protective host responses remains a research priority. Understanding the molecular and immunological mechanisms of the crosstalk between the parasite and the host is a prerequisite for the rational discovery and development of a safe, affordable, and protective anti-malaria vaccine.
Collapse
Affiliation(s)
- Julius Clemence Hafalla
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| | | | | |
Collapse
|
49
|
Schmidt NW, Butler NS, Harty JT. Plasmodium-host interactions directly influence the threshold of memory CD8 T cells required for protective immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:5873-84. [PMID: 21460205 PMCID: PMC3087867 DOI: 10.4049/jimmunol.1100194] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Plasmodium infections are responsible for millions of cases of malaria and ∼1 million deaths annually. Recently, we showed that sterile protection (95%) in BALB/c mice required Plasmodium berghei circumsporozoite protein (CS(252-260))-specific memory CD8 T cells exceeding a threshold of 1% of all PBLs. Importantly, it is not known if Plasmodium species affect the threshold of CS-specific memory CD8 T cells required for protection. Furthermore, C57BL/6 mice immunized with radiation-attenuated parasites are more difficult to protect against Plasmodium sporozoite challenge than similarly immunized BALB/c mice; however, it is not known whether this is the result of different CD8 T cell specificity, functional attributes of CD8 T cells, or mouse strain-specific factors expressed in nonhematopoietic cells. In this article, we show that more CS-specific memory CD8 T cells are required for protection against P. yoelii sporozoite challenge than for protection against P. berghei sporozoite challenge. Furthermore, P. berghei CS(252)-specific CD8 T cells exhibit reduced protection against P. berghei sporozoite challenge in the context of C57BL/6 and C57BL/10 non-MHC-linked genes in CB6F1 and B10.D2 mice, respectively. Generation and immunization of reciprocal chimeric mice between BALB/c and B10.D2 strains revealed that B10 background factors expressed by nonhematopoietic cells increased the threshold required for protection through a CD8 T cell-extrinsic mechanism. Finally, reduced CS-specific memory CD8 T cell protection in P. yoelii-infected BALB/c or P. berghei-infected B10.D2 mice correlated with increased rates of Plasmodium amplification in the liver. Thus, both Plasmodium species and strain-specific background genes in nonhematopoietic cells determine the threshold of memory CD8 T cells required for protection.
Collapse
Affiliation(s)
| | - Noah S. Butler
- Department of Microbiology, University of Iowa, Iowa City, IA, USA
| | - John T. Harty
- Department of Microbiology, University of Iowa, Iowa City, IA, USA
- Department of Pathology, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
50
|
Malaria Vaccine Development: Are Bacterial Flagellin Fusion Proteins the Bridge between Mouse and Humans? J Parasitol Res 2011; 2011:965369. [PMID: 21603205 PMCID: PMC3095412 DOI: 10.1155/2011/965369] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 01/18/2011] [Indexed: 12/25/2022] Open
Abstract
In the past 25 years, the development of an effective malaria vaccine has become one of the biggest riddles in the biomedical sciences. Experimental data using animal infection models demonstrated that it is possible to induce protective immunity against different stages of malaria parasites. Nonetheless, the vast body of knowledge has generated disappointments when submitted to clinical conditions and presently a single antigen formulation has progressed to the point where it may be translated into a human vaccine. In parallel, new means to increase the protective effects of antigens in general have been pursued and depicted, such as the use of bacterial flagellins as carriers/adjuvants. Flagellins activate pathways in the innate immune system of both mice and humans. The recent report of the first Phase I clinical trial of a vaccine containing a Salmonella flagellin as carrier/adjuvant may fuel the use of these proteins in vaccine formulations. Herein, we review the studies on the use of recombinant flagellins as vaccine adjuvants with malarial antigens in the light of the current state of the art of malaria vaccine development. The available information indicates that bacterial flagellins should be seriously considered for malaria vaccine formulations to the development of effective human vaccines.
Collapse
|