1
|
Cagigi A, Tinnirello R, Iannolo G, Douradinha B. Orthoflavivirus zikaense (Zika) vaccines: What are we waiting for? Int J Antimicrob Agents 2024; 64:107367. [PMID: 39490448 DOI: 10.1016/j.ijantimicag.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Alberto Cagigi
- International Vaccine Institute (IVI) Europe Regional Office, Solna, Sweden
| | | | | | - Bruno Douradinha
- Vaccine Technology Subgroup, Emerging Pathogens Group, Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
Ooi EE, Kalimuddin S. Lessons for dengue vaccines from a human challenge study. THE LANCET. INFECTIOUS DISEASES 2024; 24:801-803. [PMID: 38679034 DOI: 10.1016/s1473-3099(24)00153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 05/01/2024]
Affiliation(s)
- Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore; Department of Translational Clinical Research, Singapore General Hospital, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore.
| | - Shirin Kalimuddin
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Infectious Diseases, Singapore General Hospital, Singapore
| |
Collapse
|
3
|
Lyke KE, Chua JV, Koren M, Friberg H, Gromowski GD, Rapaka RR, Waickman AT, Joshi S, Strauss K, McCracken MK, Gutierrez-Barbosa H, Shrestha B, Culbertson C, Bernal P, De La Barrera RA, Currier JR, Jarman RG, Edelman R. Efficacy and immunogenicity following dengue virus-1 human challenge after a tetravalent prime-boost dengue vaccine regimen: an open-label, phase 1 trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:896-908. [PMID: 38679035 DOI: 10.1016/s1473-3099(24)00100-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/26/2024] [Accepted: 02/08/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Dengue human infection models (DHIMs) are important tools to down-select dengue vaccine candidates and establish tetravalent efficacy before advanced clinical field trials. We aimed to provide data for the safety and immunogenicity of DHIM and evaluate dengue vaccine efficacy. METHODS We performed an open-label, phase 1 trial at the University of Maryland (Baltimore, MD, USA). Eligible participants were healthy individuals aged 18-50 years who either previously received a tetravalent dengue purified inactivated vaccine prime followed by a live-attenuated vaccine boost (ie, the vaccinee group), or were unvaccinated flavivirus-naive participants (ie, the control group). Participants in the vaccinee group with detectable pre-challenge dengue virus-1 neutralising antibody titres and flavivirus-naive participants in the control group were inoculated with dengue virus-1 strain 45AZ5 in the deltoid region, 27-65 months following booster dosing. These participants were followed-up from days 4-16 following dengue virus-1 live virus human challenge, with daily real-time quantitative PCR specific to dengue virus-1 RNA detection, and dengue virus-1 solicited local and systemic adverse events were recorded. The primary outcomes were safety (ie, solicited local and systemic adverse events) and vaccine efficacy (ie, dengue virus-1 RNAaemia) following dengue challenge. This study is registered with ClinicalTrials.gov, number NCT04786457. FINDINGS In January 2021, ten eligible participants were enrolled; of whom, six (60%) were in the vaccinee group and four (40%) were in the control group. Daily quantitative PCR detected dengue virus-1 RNA in nine (90%) of ten participants (five [83%] of six in the vaccinee group and all four [100%] in the control group). The mean onset of RNAaemia occurred on day 5 (SD 1·0) in the vaccinee group versus day 8 (1·5) in the control group (95% CI 1·1-4·9; p=0·007), with a trend towards reduced RNAaemia duration in the vaccinee group compared with the control group (8·2 days vs 10·5 days; 95% CI -0·08 to 4·68; p=0·056). Mild-to-moderate symptoms (nine [90%] of ten), leukopenia (eight [89%] of nine), and elevated aminotransferases (seven [78%] of nine) were commonly observed. Severe adverse events were detected only in the vaccinee group (fever ≥38·9°C in three [50%] of six, headache in one [17%], and transient grade 4 aspartate aminotransferase elevation in one [17%]). No deaths were reported. INTERPRETATION Participants who had tetravalent dengue purified inactivated vaccine prime and live-attenuated vaccine boost were unprotected against dengue virus-1 infection and further showed increased clinical, immunological, and transcriptomic evidence for inflammation potentially mediated by pre-existing infection-enhancing antibodies. This study highlights the impact of small cohort, human challenge models studying dengue pathogenesis and downstream vaccine development. FUNDING Military Infectious Disease Research Program and Medical Technology Enterprise Consortium and Advanced Technology International.
Collapse
Affiliation(s)
- Kirsten E Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Joel V Chua
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael Koren
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Heather Friberg
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gregory D Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adam T Waickman
- Department of Microbiology and Immunology, State University of New York Update Medical University, Syracuse, NY, USA
| | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathleen Strauss
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael K McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher Culbertson
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Paula Bernal
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rafael A De La Barrera
- Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Richard G Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Robert Edelman
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Waickman AT, Newell K, Lu JQ, Fang H, Waldran M, Gebo C, Currier JR, Friberg H, Jarman RG, Klick MD, Ware LA, Endy TP, Thomas SJ. Low-dose dengue virus 3 human challenge model: a phase 1 open-label study. Nat Microbiol 2024; 9:1356-1367. [PMID: 38561497 DOI: 10.1038/s41564-024-01668-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024]
Abstract
Dengue human infection models present an opportunity to explore the potential of a vaccine, anti-viral or immuno-compound for clinical benefit in a controlled setting. Here we report the outcome of a phase 1 open-label assessment of a low-dose dengue virus 3 (DENV-3) challenge model (NCT04298138), in which nine participants received a subcutaneous inoculation with 0.5 ml of a 1.4 × 103 plaque-forming unit per ml suspension of the attenuated DENV-3 strain CH53489. The primary and secondary endpoints of the study were to assess the safety of this DENV-3 strain in healthy flavivirus-seronegative individuals. All participants developed RNAaemia within 7 days after inoculation with peak titre ranging from 3.13 × 104 to 7.02 × 108 genome equivalents per ml. Solicited symptoms such as fever and rash, clinical laboratory abnormalities such as lymphopenia and thrombocytopenia, and self-reported symptoms such as myalgia were consistent with mild-to-moderate dengue in all volunteers. DENV-3-specific seroconversion and memory T cell responses were observed within 14 days after inoculation as assessed by enzyme-linked immunosorbent assay and interferon-gamma-based enzyme-linked immunospot. RNA sequencing and serum cytokine analysis revealed anti-viral responses that overlapped with the period of viraemia. The magnitude and frequency of clinical and immunologic endpoints correlated with an individual's peak viral titre.
Collapse
Affiliation(s)
- Adam T Waickman
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA.
- Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, USA.
| | - Krista Newell
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Joseph Q Lu
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - HengSheng Fang
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Mitchell Waldran
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Chad Gebo
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Heather Friberg
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Richard G Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Michelle D Klick
- Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Lisa A Ware
- Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Timothy P Endy
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA.
- Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
5
|
Oxford JS, Catchpole A, Mann A, Bell A, Noulin N, Gill D, Oxford JR, Gilbert A, Balasingam S. A Brief History of Human Challenge Studies (1900-2021) Emphasising the Virology, Regulatory and Ethical Requirements, Raison D'etre, Ethnography, Selection of Volunteers and Unit Design. Curr Top Microbiol Immunol 2024; 445:1-32. [PMID: 35704095 DOI: 10.1007/82_2022_253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Venetian quarantine 400 years ago was an important public health measure. Since 1900 this has been refined to include "challenge" or deliberate infection with pathogens be they viruses, bacteria, or parasites. Our focus is virology and ranges from the early experiments in Cuba with Yellow Fever Virus to the most widespread pathogen of our current times, COVID-19. The latter has so far caused over four million deaths worldwide and 190 million cases of the disease. Quarantine and challenge were also used to investigate the Spanish Influenza of 1918 which caused over 100 million deaths. We consider here the merits of the approach, that is the speeding up of knowledge in a practical sense leading to the more rapid licensing of vaccines and antimicrobials. At the core of quarantine and challenge initiatives is the design of the unit to allow safe confinement of the pathogen and protection of the staff. Most important though is the safety of volunteers. We can see now, as in 1900, that members of our society are prepared and willing to engage in these experiments for the public good. Our ethnology study, where the investigator observed the experiment from within the quarantine, gave us the first indication of changing attitudes amongst volunteers whilst in quarantine. These quarantine experiments, referred to as challenge studies, human infection studies, or "controlled human infection models" involve thousands of clinical samples taken over two to three weeks and can provide a wealth of immunological and molecular data on the infection itself and could allow the discovery of new targets for vaccines and therapeutics. The Yellow Fever studies from 121 years ago gave the impetus for development of a successful vaccine still used today whilst also uncovering the nature of the Yellow Fever agent, namely that it was a virus. We outline how carefully these experiments are approached and the necessity to have high quality units with self-contained air-flow along with extensive personal protective equipment for nursing and medical staff. Most important is the employment of highly trained scientific, medical and nursing staff. We face a future of emerging pathogens driven by the increasing global population, deforestation, climate change, antibiotic resistance and increased global travel. These emerging pathogens may be pathogens we currently are not aware of or have not caused outbreaks historically but could also be mutated forms of known pathogens including viruses such as influenza (H7N9, H5N1 etc.) and coronaviruses. This calls for challenge studies to be part of future pandemic preparedness as an additional tool to assist with the rapid development of broad-spectrum antimicrobials, immunomodulators and new vaccines.
Collapse
Affiliation(s)
- J S Oxford
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | | | | | | | | | - D Gill
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | - J R Oxford
- Inveresk Medical Practice, Edinburgh, E21 7BP, UK
| | | | | |
Collapse
|
6
|
Ooi EE, Kalimuddin S. Insights into dengue immunity from vaccine trials. Sci Transl Med 2023; 15:eadh3067. [PMID: 37437017 DOI: 10.1126/scitranslmed.adh3067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The quest for an effective dengue vaccine has culminated in two approved vaccines and another that has completed phase 3 clinical trials. However, shortcomings exist in each, suggesting that the knowledge on dengue immunity used to develop these vaccines was incomplete. Vaccine trial findings could refine our understanding of dengue immunity, because these are experimentally derived, placebo-controlled data. Results from these trials suggest that neutralizing antibody titers alone are insufficient to inform protection against symptomatic infection, implicating a role for cellular immunity in protection. These findings have relevance for both future dengue vaccine development and application of current vaccines for maximal public health benefit.
Collapse
Affiliation(s)
- Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore 169857, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Shirin Kalimuddin
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore 169856, Singapore
| |
Collapse
|
7
|
Odio CD, Lowman KE, Law M, Aogo RA, Hunsberger S, Wood BJ, Kassin M, Levy E, Callier V, Firdous S, Hasund CM, Voirin C, Kattappuram R, Yek C, Manning J, Durbin A, Whitehead SS, Katzelnick LC. Phase 1 trial to model primary, secondary, and tertiary dengue using a monovalent vaccine. BMC Infect Dis 2023; 23:345. [PMID: 37221466 DOI: 10.1186/s12879-023-08299-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND The four co-circulating and immunologically interactive dengue virus serotypes (DENV1-4) pose a unique challenge to vaccine design because sub-protective immunity can increase the risk of severe dengue disease. Existing dengue vaccines have lower efficacy in DENV seronegative individuals but higher efficacy in DENV exposed individuals. There is an urgent need to identify immunological measures that are strongly associated with protection against viral replication and disease following sequential exposure to distinct serotypes. METHODS/DESIGN This is a phase 1 trial wherein healthy adults with neutralizing antibodies to zero (seronegative), one non-DENV3 (heterotypic), or more than one (polytypic) DENV serotype will be vaccinated with the live attenuated DENV3 monovalent vaccine rDEN3Δ30/31-7164. We will examine how pre-vaccine host immunity influences the safety and immunogenicity of DENV3 vaccination in a non-endemic population. We hypothesize that the vaccine will be safe and well tolerated, and all groups will have a significant increase in the DENV1-4 neutralizing antibody geometric mean titer between days 0 and 28. Compared to the seronegative group, the polytypic group will have lower mean peak vaccine viremia, due to protection conferred by prior DENV exposure, while the heterotypic group will have higher mean peak viremia, due to mild enhancement. Secondary and exploratory endpoints include characterizing serological, innate, and adaptive cell responses; evaluating proviral or antiviral contributions of DENV-infected cells; and immunologically profiling the transcriptome, surface proteins, and B and T cell receptor sequences and affinities of single cells in both peripheral blood and draining lymph nodes sampled via serial image-guided fine needle aspiration. DISCUSSION This trial will compare the immune responses after primary, secondary, and tertiary DENV exposure in naturally infected humans living in non-endemic areas. By evaluating dengue vaccines in a new population and modeling the induction of cross-serotypic immunity, this work may inform vaccine evaluation and broaden potential target populations. TRIAL REGISTRATION NCT05691530 registered on January 20, 2023.
Collapse
Affiliation(s)
- Camila D Odio
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Kelsey E Lowman
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Melissa Law
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rosemary A Aogo
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sally Hunsberger
- Division of Clinical Research, Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brad J Wood
- Interventional Radiology and Center for Interventional Oncology, NIH Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Kassin
- Interventional Radiology and Center for Interventional Oncology, NIH Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elliot Levy
- Interventional Radiology and Center for Interventional Oncology, NIH Clinical Center and National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Viviane Callier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Saba Firdous
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chloe M Hasund
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charlie Voirin
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robbie Kattappuram
- Department of Pharmacy, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Christina Yek
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jessica Manning
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anna Durbin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stephen S Whitehead
- Arbovirus Vaccine Research Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Waickman AT, Newell K, Endy TP, Thomas SJ. Biologics for dengue prevention: up-to-date. Expert Opin Biol Ther 2023; 23:73-87. [PMID: 36417290 DOI: 10.1080/14712598.2022.2151837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Dengue is a worsening global public health problem. The vector-viral-host interactions driving the pathogenesis of dengue are multi-dimensional. Sequential dengue virus (DENV) infections with different DENV types significantly increase the risk of severe disease. Treatment is supportive in nature as there are no licensed anti-DENV antivirals or immuno-therapeutics. A single dengue vaccine has widely been licensed with two others in advanced clinical development. Dengvaxia® has been licensed in numerous countries but uptake has been slow as a result of safety signals noted in the youngest recipients and those who were dengue naïve at the time of vaccination. AREAS COVERED In this review, the current state of dengue vaccine and antiviral drug development will be discussed as well as new developments in controlled human infection models to support product development. EXPERT OPINION The world needs a safe and efficacious tetravalent dengue vaccine capable of protecting multiple different populations across a broad age range and different flavivirus immunologic backgrounds. Safe and effective antivirals are also needed to prevent or attenuate dengue disease in the unvaccinated, in cases of vaccine failure, or in high-risk populations.
Collapse
Affiliation(s)
- Adam T Waickman
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| | - Krista Newell
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| | - Timothy P Endy
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| |
Collapse
|
9
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
10
|
Idris F, Ting DHR, Alonso S. An update on dengue vaccine development, challenges, and future perspectives. Expert Opin Drug Discov 2021. [DOI: 10.1080/17460441.2020.1811675
expr 880867630 + 907120263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Affiliation(s)
- Fakhriedzwan Idris
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Donald Heng Rong Ting
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sylvie Alonso
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
King CA, Wegman AD, Endy TP. Mobilization and Activation of the Innate Immune Response to Dengue Virus. Front Cell Infect Microbiol 2020; 10:574417. [PMID: 33224897 PMCID: PMC7670994 DOI: 10.3389/fcimb.2020.574417] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022] Open
Abstract
Dengue virus is an important human pathogen, infecting an estimated 400 million individuals per year and causing symptomatic disease in a subset of approximately 100 million. Much of the effort to date describing the host response to dengue has focused on the adaptive immune response, in part because of the well-established roles of antibody-dependent enhancement and T cell original sin as drivers of severe dengue upon heterotypic secondary infection. However, the innate immune system is a crucial factor in the host response to dengue, as it both governs the fate and vigor of the adaptive immune response, and mediates the acute inflammatory response in tissues. In this review, we discuss the innate inflammatory response to dengue infection, focusing on the role of evolutionarily conserved innate immune cells, their effector functions, and clinical course.
Collapse
Affiliation(s)
- Christine A. King
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | | | | |
Collapse
|
12
|
Idris F, Ting DHR, Alonso S. An update on dengue vaccine development, challenges, and future perspectives. Expert Opin Drug Discov 2020; 16:47-58. [PMID: 32838577 DOI: 10.1080/17460441.2020.1811675] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION From both a public health and economic perspective, vaccination is arguably the most effective approach to combat endemic and pandemic infectious diseases. Dengue affects more than 100 countries in the tropical and subtropical world, with 100-400 million infections every year. In the wake of the recent setback faced by Dengvaxia, the only FDA-approved dengue vaccine, safer and more effective dengue vaccines candidates are moving along the clinical pipeline. AREA COVERED This review provides an update of the latest outcomes of dengue vaccine clinical trials. In the light of recent progress made in our understanding of dengue pathogenesis and immune correlates of protection, novel vaccine strategies have emerged with promising second-generation dengue vaccine candidates. Finally, the authors discuss the dengue-specific challenges that remain to be addressed and overcome. EXPERT OPINION The authors propose to explore various adjuvants and delivery systems that may help improve the design of safe, effective, and affordable vaccines against dengue. They also challenge the concept of a 'universal' dengue vaccine as increasing evidence support that DENV strains have evolved different virulence mechanisms.
Collapse
Affiliation(s)
- Fakhriedzwan Idris
- Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore , Singapore, Singapore
| | - Donald Heng Rong Ting
- Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore , Singapore, Singapore
| | - Sylvie Alonso
- Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore , Singapore, Singapore
| |
Collapse
|
13
|
Bekeredjian-Ding I, Van Molle W, Baay M, Neels P. Human challenge trial workshop: Focus on quality requirements for challenge agents, Langen, Germany, October 22, 2019. Biologicals 2020; 66:53-61. [PMID: 32389512 DOI: 10.1016/j.biologicals.2020.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/01/2023] Open
Abstract
Controlled human infection models can be helpful to study pathogenesis and immune responses as a basis for the development of vaccines. In controlled human infection models, human challenge agents are used to infect healthy volunteers, therefore, ethical considerations include that the exposure studies need to be safe and results should be meaningful, e.g. contribute to a better cure. Both in the US and in Europe, the level of Good Manufacturing Practice required is related to the phase of the study ('sliding scale Good Manufacturing Practice'), and, hence, is much more open to speedy drug development than anticipated. Recommendations included: the development of guidelines for human challenge agents; a focus on strain selection, in particular with regard to strain infectivity, stability and purity; the use of whole genome sequencing; a reference repository of challenge agents, the need for early exchange with regulators to ensure acceptability of strain selection and manufacturing for later drug development; sharing of models and challenge agents.
Collapse
Affiliation(s)
- Isabelle Bekeredjian-Ding
- Paul-Ehrlich-Institut (PEI), Langen, Germany; Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.
| | | | - Marc Baay
- P95 Epidemiology & Pharmacovigilance, Leuven, Belgium.
| | - Pieter Neels
- International Alliance for Biological Standardization (IABS), Belgium.
| | | |
Collapse
|
14
|
Route of inoculation and mosquito vector exposure modulate dengue virus replication kinetics and immune responses in rhesus macaques. PLoS Negl Trop Dis 2020; 14:e0008191. [PMID: 32267846 PMCID: PMC7141610 DOI: 10.1371/journal.pntd.0008191] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/02/2020] [Indexed: 02/04/2023] Open
Abstract
Dengue virus (DENV) is transmitted by infectious mosquitoes during blood-feeding via saliva containing biologically-active proteins. Here, we examined the effect of varying DENV infection modality in rhesus macaques in order to improve the DENV nonhuman primate (NHP) challenge model. NHPs were exposed to DENV-1 via subcutaneous or intradermal inoculation of virus only, intradermal inoculation of virus and salivary gland extract, or infectious mosquito feeding. The infectious mosquito feeding group exhibited delayed onset of viremia, greater viral loads, and altered clinical and immune responses compared to other groups. After 15 months, NHPs in the subcutaneous and infectious mosquito feeding groups were re-exposed to either DENV-1 or DENV-2. Viral replication and neutralizing antibody following homologous challenge were suggestive of sterilizing immunity, whereas heterologous challenge resulted in productive, yet reduced, DENV-2 replication and boosted neutralizing antibody. These results show that a more transmission-relevant exposure modality resulted in viral replication closer to that observed in humans. Dengue virus is transmitted into the skin of humans by mosquitoes as they take a blood meal. In contrast, many animal models are infected in the laboratory using a syringe to inject below the skin. Here, we looked at how different routes and methods of infection altered dengue infection in rhesus macaques. We found that infection via mosquito feeding resulted in a number of changes compared to other routes and methods, including a delay in the time to detection of dengue virus and overall greater quantities of dengue virus in the blood, and changes in the amounts of various components of blood that have been associated with dengue disease in humans. After 15 months, we exposed the macaques again to either the same or a different type of dengue virus. We found that animals exposed to the same type of dengue virus were protected from infection, whereas those animals exposed to a different type were only partially protected. Overall, our results show that dengue virus delivery using the natural transmission vector, mosquitoes, results in infection that is closer to what is observed in humans and may influence the interpretation of future studies of candidate vaccines.
Collapse
|
15
|
Non-Human Primate Models of Dengue Virus Infection: A Comparison of Viremia Levels and Antibody Responses during Primary and Secondary Infection among Old World and New World Monkeys. Pathogens 2020; 9:pathogens9040247. [PMID: 32230836 PMCID: PMC7238212 DOI: 10.3390/pathogens9040247] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022] Open
Abstract
Due to the global burden of dengue disease, a vaccine is urgently needed. One of the key points in vaccine development is the development of a robust and reliable animal model of dengue virus infection. Characteristics including the ability to sustain viral replication, demonstration of clinical signs, and immune response that resemble those of human dengue virus infection are vital in animal models. Preclinical studies in vaccine development usually include parameters such as safety evaluation, induction of viremia and antigenemia, immunogenicity, and vaccine effectiveness. Although mice have been used as a model, non-human primates have an advantage over mice because of their relative similarity to humans in their genetic composition and immune responses. This review compares the viremia kinetics and antibody responses of cynomolgus macaques (Macaca fasicularis), common marmosets (Callithrix jacchus), and tamarins (Saguinus midas and Saguinus labitus) and summarize the perspectives and the usefulness along with challenges in dengue vaccine development.
Collapse
|
16
|
Hooft van Huijsduijnen R, Kojima S, Carter D, Okabe H, Sato A, Akahata W, Wells TNC, Katsuno K. Reassessing therapeutic antibodies for neglected and tropical diseases. PLoS Negl Trop Dis 2020; 14:e0007860. [PMID: 31999695 PMCID: PMC6991954 DOI: 10.1371/journal.pntd.0007860] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In the past two decades there has been a significant expansion in the number of new therapeutic monoclonal antibodies (mAbs) that are approved by regulators. The discovery of these new medicines has been driven primarily by new approaches in inflammatory diseases and oncology, especially in immuno-oncology. Other recent successes have included new antibodies for use in viral diseases, including HIV. The perception of very high costs associated with mAbs has led to the assumption that they play no role in prophylaxis for diseases of poverty. However, improvements in antibody-expression yields and manufacturing processes indicate this is a cost-effective option for providing protection from many types of infection that should be revisited. Recent technology developments also indicate that several months of protection could be achieved with a single dose. Moreover, new methods in B cell sorting now enable the systematic identification of high-quality antibodies from humanized mice, or patients. This Review discusses the potential for passive immunization against schistosomiasis, fungal infections, dengue, and other neglected diseases.
Collapse
Affiliation(s)
| | | | - Dee Carter
- School of Life and Environmental Sciences and The Marie Bashir Institute, University of Sydney, NSW, Australia
| | | | | | - Wataru Akahata
- VLP Therapeutics, Gaithersburg, Maryland, United States of America
| | | | - Kei Katsuno
- Global Health Innovative Technology Fund, Tokyo, Japan
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Nagasaki University School of Tropical Medicine and Global Health, Nagasaki, Japan
| |
Collapse
|
17
|
Meeting Report: WHO consultation on considerations for regulatory expectations of Zika virus vaccines for use during an emergency. Vaccine 2019; 37:7443-7450. [DOI: 10.1016/j.vaccine.2016.10.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 12/25/2022]
|
18
|
Gamma Radiation Sterilization Dose of Adult Males in Asian Tiger Mosquito Pupae. INSECTS 2019; 10:insects10040101. [PMID: 30965594 PMCID: PMC6523977 DOI: 10.3390/insects10040101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 11/17/2022]
Abstract
The pathogen-carrying tiger mosquito, Aedes albopictus, has spread from the Western Pacific and Southeast Asia to Europe, Africa, the Middle East, North and South America, and the Caribbean. This species of mosquito transmits arboviral infections, such as yellow fever, chikungunya, dengue, zika, and several encephalitides. The objective of this research was to provide a radiation dose inducing sterilization in adult male Ae. albopictus in the pupal stage. A cobalt-60 source of gamma radiation at a dose rate of 381 Gy/h was used. The pupae were irradiated with doses of 0 (control), 20, 30, 40, 50, and 60 Gy. Each treatment had a total of five replications using 60 pupae. After irradiation, the different phases of Ae. albopictus development (egg, larva, pupa, and adult) in the F1 generation were observed daily. Parameters such as viability, fertility, longevity, and mortality were recorded. The results from these studies showed that a dose of 60 Gy was necessary to sterilize 100% of the male Ae. albopictus pupae.
Collapse
|
19
|
Rose A, Sekhar A. Bioethics of establishing a CHIM model for dengue vaccine development. Int J Infect Dis 2019; 84S:S74-S79. [PMID: 30641207 DOI: 10.1016/j.ijid.2019.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Controlled human infection models (CHIM) have been used in vaccine development to up-select and down-select potential vaccine candidates and to provide proof of vaccine efficacy, and have also been used as a basis for licensure of vaccines for cholera and typhoid by regulatory agencies. CHIM IN DENGUE VACCINES DEVELOPMENT Dengue fever results in ∼400 million infections a year and is of significant health concern especially in India. There are currently no antivirals for the disease and the only licensed vaccine for dengue is not widely used owing to safety concerns. Controlled dengue human challenge models (DHCM) are currently being used to assess the efficacy of vaccines in development for dengue. DENGUE CHIM IN INDIA Conducting CHIM studies in India especially for evaluation of dengue vaccine candidates will be hugely beneficial as the disease is endemic to India and hence the effect of pre-exposure to the virus on vaccine safety and efficacy can be established. However, to date no CHIM studies have been conducted in India and there is a need to educate ethics committee members, policy makers and the public on the importance of such studies and what they entail.
Collapse
Affiliation(s)
- Anuradha Rose
- Departments of Community Health, Bioethics, Christian Medical College, Vellore, Tamil Nadu, India
| | - Amrita Sekhar
- Departments of Community Health, Bioethics, Christian Medical College, Vellore, Tamil Nadu, India.
| |
Collapse
|
20
|
Thomas SJ, Agulto L, Hendrickx K, Erpicum M, Tomashek KM, Cassetti MC, Laughlin C, Precioso A, Schmidt AC, Narvaez F, Siqueira JB, Tissera H, Edelman R. Dengue illness index-A tool to characterize the subjective dengue illness experience. PLoS Negl Trop Dis 2018; 12:e0006593. [PMID: 30286086 PMCID: PMC6171819 DOI: 10.1371/journal.pntd.0006593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 06/08/2018] [Indexed: 02/04/2023] Open
Abstract
Dengue virus infections are a major cause of febrile illness that significantly affects individual and societal productivity and drives up health care costs principally in the developing world. Two dengue vaccine candidates are in advanced clinical efficacy trials in Latin America and Asia, and another has been licensed in more than fifteen countries but its uptake has been limited. Despite these advances, standardized metrics for comparability of protective efficacy between dengue vaccines remain poorly defined. The Dengue Illness Index (DII) is a tool that we developed thru refinement of previous similar iterations in an attempt to improve and standardize the measurement of vaccine and drug efficacy in reducing moderate dengue illness. The tool is designed to capture an individual's overall disease experience based on how the totality of their symptoms impacts their general wellness and daily functionality. We applied the DII to a diary card, the Dengue Illness Card (DIC), which was examined and further developed by a working group. The card was then refined with feedback garnered from a Delphi methodology-based query that addressed the adequacy and applicability of the tool in clinical dengue research. There was overall agreement that the tool would generate useful data and provide an alternative perspective to the assessment of drug or vaccine candidates, which in the case of vaccines, are assessed by their reduction in any virologically confirmed dengue of any severity with a focus on the more severe. The DIC needs to be evaluated in the field in the context of vaccine or drug trials, prospective cohort studies, or during experimental human infection studies. Here, we present the final DIC resulting from the Delphi process and offer its further development or use to the dengue research community.
Collapse
Affiliation(s)
- Stephen J. Thomas
- Division of Infectious Diseases, State University of New York, Upstate Medical University, Syracuse, NY, United States of America
- * E-mail:
| | - Liane Agulto
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States of America
| | - Kim Hendrickx
- FWO Postdoctoral Fellow of the Life Sciences & Society Lab, Center for Sociological Research, KU Leuven, Flanders, Belgium
- SPIRAL Research Center, Département de Science Politique, Université de Liège, Liège, Belgium
| | - Martin Erpicum
- SPIRAL Research Center, Département de Science Politique, Université de Liège, Liège, Belgium
| | - Kay M. Tomashek
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States of America
| | - M. Cristina Cassetti
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States of America
| | - Catherine Laughlin
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States of America
| | - Alexander Precioso
- Division of Clinical Trials and Pharmacovigilance, Butantan Institute, São Paulo, Brazil
- Pediatrics Department of the Medical School of University of São Paulo, São Paulo, Brazil
| | | | - Federico Narvaez
- Infectious Diseases Unit, National Pediatric Reference Hospital, Hospital Infantil Manuel de Jesús Rivera, Managua, Nicaragua
| | | | - Hasitha Tissera
- National Dengue Control Unit, Ministry of Health, Colombo, Sri Lanka
| | - Robert Edelman
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
21
|
Vannice KS, Wilder-Smith A, Barrett ADT, Carrijo K, Cavaleri M, de Silva A, Durbin AP, Endy T, Harris E, Innis BL, Katzelnick LC, Smith PG, Sun W, Thomas SJ, Hombach J. Clinical development and regulatory points for consideration for second-generation live attenuated dengue vaccines. Vaccine 2018; 36:3411-3417. [PMID: 29525283 PMCID: PMC6010224 DOI: 10.1016/j.vaccine.2018.02.062] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/05/2018] [Accepted: 02/15/2018] [Indexed: 01/05/2023]
Abstract
Licensing and decisions on public health use of a vaccine rely on a robust clinical development program that permits a risk-benefit assessment of the product in the target population. Studies undertaken early in clinical development, as well as well-designed pivotal trials, allow for this robust characterization. In 2012, WHO published guidelines on the quality, safety and efficacy of live attenuated dengue tetravalent vaccines. Subsequently, efficacy and longer-term follow-up data have become available from two Phase 3 trials of a dengue vaccine, conducted in parallel, and the vaccine was licensed in December 2015. The findings and interpretation of the results from these trials released both before and after licensure have highlighted key complexities for tetravalent dengue vaccines, including concerns vaccination could increase the incidence of dengue disease in certain subpopulations. This report summarizes clinical and regulatory points for consideration that may guide vaccine developers on some aspects of trial design and facilitate regulatory review to enable broader public health recommendations for second-generation dengue vaccines.
Collapse
Affiliation(s)
- Kirsten S Vannice
- World Health Organization, Department of Immunizations, Vaccines and Biologicals, Geneva, Switzerland
| | - Annelies Wilder-Smith
- World Health Organization, Department of Immunizations, Vaccines and Biologicals, Geneva, Switzerland; Lee Kong Chian School of Medicine, Singapore
| | - Alan D T Barrett
- Sealy Center for Vaccine Development and World Health Organization Collaborating Center for Vaccine Research, Evaluation and Training for Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Kalinka Carrijo
- Brazilian Health Regulatory Agency - Anvisa, Brasília, DF, Brazil
| | | | - Aravinda de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Anna P Durbin
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Tim Endy
- State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Bruce L Innis
- Respiratory Infections and Maternal Immunizations, PATH Center for Vaccine Innovation and Access, Washington, DC, USA
| | - Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Peter G Smith
- Tropical Epidemiology Group, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Stephen J Thomas
- State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Joachim Hombach
- World Health Organization, Department of Immunizations, Vaccines and Biologicals, Geneva, Switzerland.
| |
Collapse
|
22
|
Experimental in vitro and in vivo systems for studying the innate immune response during dengue virus infections. Arch Virol 2018. [PMID: 29520688 DOI: 10.1007/s00705-018-3784-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Dengue is the most prevalent arboviral disease in humans and leads to significant morbidity and socioeconomic burden in tropical and subtropical areas. Dengue is caused by infection with any of the four closely related serotypes of dengue virus (DENV1-4) and usually manifests as a mild febrile illness, but may develop into fatal dengue hemorrhagic fever and shock syndrome. There are no specific antiviral therapies against dengue because understanding of DENV biology is limited. A tetravalent chimeric dengue vaccine, Dengvaxia, has finally been licensed for use, but its efficacy was significantly lower against DENV-2 infections and in dengue-naïve individuals. The identification of mechanisms underlying the interactions between DENV and immune responses will help to determine efficient therapeutic and preventive options. It has been well established how the innate immune system responds to DENV infection and how DENV overcomes innate antiviral defenses, however further progress in this field remains hampered by the absence of appropriate experimental dengue models. Herein, we review the available in vitro and in vivo approaches to study the innate immune responses to DENV.
Collapse
|
23
|
Züst R, Li SH, Xie X, Velumani S, Chng M, Toh YX, Zou J, Dong H, Shan C, Pang J, Qin CF, Newell EW, Shi PY, Fink K. Characterization of a candidate tetravalent vaccine based on 2'-O-methyltransferase mutants. PLoS One 2018; 13:e0189262. [PMID: 29298302 PMCID: PMC5751980 DOI: 10.1371/journal.pone.0189262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/23/2017] [Indexed: 11/26/2022] Open
Abstract
Dengue virus (DENV) is one of the most widespread arboviruses. The four DENV serotypes infect about 400 million people every year, causing 96 million clinical dengue cases, of which approximately 500’000 are severe and potentially life-threatening. The only licensed vaccine has a limited efficacy and is only recommended in regions with high endemicity. We previously reported that 2’-O-methyltransferase mutations in DENV-1 and DENV-2 block their capacity to inhibit type I IFNs and render the viruses attenuated in vivo, making them amenable as vaccine strains; here we apply this strategy to all four DENV serotypes to generate a tetravalent, non-chimeric live-attenuated dengue vaccine. 2’-O-methyltransferase mutants of all four serotypes are highly sensitive to type I IFN inhibition in human cells. The tetravalent formulation is attenuated and immunogenic in mice and cynomolgus macaques and elicits a response that protects from virus challenge. These results show the potential of 2’-O-methyltransferase mutant viruses as a safe, tetravalent, non-chimeric dengue vaccine.
Collapse
Affiliation(s)
- Roland Züst
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Shi-Hua Li
- Novartis Institute for Tropical Diseases, Chromos, Singapore, Singapore
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xuping Xie
- Novartis Institute for Tropical Diseases, Chromos, Singapore, Singapore
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Sumathy Velumani
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Melissa Chng
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ying-Xiu Toh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jing Zou
- Novartis Institute for Tropical Diseases, Chromos, Singapore, Singapore
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Hongping Dong
- Novartis Institute for Tropical Diseases, Chromos, Singapore, Singapore
| | - Chao Shan
- Novartis Institute for Tropical Diseases, Chromos, Singapore, Singapore
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Jassia Pang
- Biological Resource Centre, Agency for Science, Technology and Research, Singapore, Singapore
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Evan W. Newell
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Pei-Yong Shi
- Novartis Institute for Tropical Diseases, Chromos, Singapore, Singapore
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States of America
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, United States of America
- * E-mail: (KF); (PYS)
| | - Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- * E-mail: (KF); (PYS)
| |
Collapse
|
24
|
Moi ML, Ami Y, Muhammad Azami NA, Shirai K, Yoksan S, Suzaki Y, Kitaura K, Lim CK, Saijo M, Suzuki R, Takasaki T, Kurane I. Marmosets (Callithrix jacchus) as a non-human primate model for evaluation of candidate dengue vaccines: induction and maintenance of specific protective immunity against challenges with clinical isolates. J Gen Virol 2017; 98:2955-2967. [PMID: 29160199 DOI: 10.1099/jgv.0.000913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dengue virus (DENV) is one of the major infectious diseases in tropical regions and approximately half of the world population is at risk of infection. Vaccines would offer an effective control measure against this disease. We previously reported on the utility of marmosets as an animal model for studying primary and secondary dengue infections. Infected marmosets consistently develop viraemia and antibody kinetics that reflect those of patients with dengue. Thus, it is important to determine the utility of marmosets as an animal model for demonstrating vaccine efficacy. In this study, marmosets were inoculated with candidate vaccine and parent strains and challenged with a clinical DENV strain. The viraemia and antibody kinetics in these marmosets were determined. Marmosets consistently develop lower viraemia with an attenuated vaccine strain. During secondary challenge, the IgM response was delayed, whereas the IgG levels rose rapidly, indicating a secondary antibody response. The neutralizing activities against the homotypic serotype were high; all marmosets were protected against viraemia following secondary inoculation. The viraemia markers and antibody responses were consistent with those of human DENV infection and vaccinees. These results demonstrate the utility of marmosets as an animal model for the study of vaccine efficacy.
Collapse
Affiliation(s)
- Meng Ling Moi
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Yasushi Ami
- Division of Experimental Animal Research, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | | | - Kenji Shirai
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Kanagawa, Japan
| | - Sutee Yoksan
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Yuriko Suzaki
- Division of Experimental Animal Research, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Kazutaka Kitaura
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Kanagawa, Japan
| | - Chang-Kweng Lim
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Kanagawa, Japan
| | | | - Ichiro Kurane
- National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
25
|
Aguirre-Obando OA, Martins AJ, Navarro-Silva MA. First report of the Phe1534Cys kdr mutation in natural populations of Aedes albopictus from Brazil. Parasit Vectors 2017; 10:160. [PMID: 28347326 PMCID: PMC5369189 DOI: 10.1186/s13071-017-2089-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 03/15/2017] [Indexed: 11/10/2022] Open
Abstract
Background Knockdown resistance (kdr), caused by alterations in the voltage-gated sodium channel (NaV), is one of the mechanisms responsible for pyrethroid (PY) resistance. In the Asian tiger mosquito, Aedes albopictus, at least four different mutations were described in the IIIS6 NaV segment in populations from Asia, North America and Europe. In contrast, in Aedes aegypti at least 12 non-synonymous mutations have been reported at nine different codons, mostly in the IIS6 and IIIS6 NaV segments. The Phe1534Cys kdr mutation in the IIIS6 NaV segment is the most prevalent in populations of Ae. aegypti worldwide, also found in Ae. albopictus from Singapore. Herein, we investigated the DNA diversity corresponding to the IIS6 and IIIS6 NaV segments in natural populations of Ae. albopictus from Brazil. Methods DNA from eight Brazilian Ae. albopictus natural populations were individually extracted and pooled by states of origin, amplified, cloned and sequenced for the corresponding IIS6 and IIIS6 NaV segments. Additionally, samples from each location were individually genotyped by an allelic specific PCR (AS-PCR) approach to obtain the genotypic and allelic frequencies for the 1534 NaV site. Results No non-synonymous substitutions were observed in the IIS6 sequences. However, the Phe1534Cys kdr mutation was evidenced in the Ae. albopictus NaV IIIS6 segment sequences from Paraná (PR) and Rondônia (RO) states, but not from Mato Grosso (MT) state. The 1534Cyskdr allele varied from 3% (Marilena/PR and Porto Velho/RO) to 10% (Foz do Iguaçu/PR). To our knowledge, this paper reports the first occurrence and provides distribution data of a possible kdr mutation in Ae. albopictus in South America. Conclusion The emergence of a likely kdr mutation in Ae. albopitus natural populations is a signal of alert for vector control measures since PY are the most popular insecticides adopted by residents. Additionally, once the kdr allele is present, its frequency tends to increase faster under exposition to those compounds. Although the Asian tiger mosquito is not incriminated as an important vector of dengue, chikungunya and Zika viruses in South America, its importance in this regard has been extensively discussed since Ae. albopictus is rapidly spreading and can also migrate between sylvatic and urban environments. Therefore, insecticide resistance monitoring initiatives should also be extended to Ae. albopictus in Brazil in order to maintain chemical compounds as an efficient vector control tool when needed. Electronic supplementary material The online version of this article (doi:10.1186/s13071-017-2089-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Oscar Alexander Aguirre-Obando
- Laboratório de Entomologia Médica e Veterinária, Setor de Ciências Biológicas, Departamento de Zoologia, Universidade Federal do Paraná, PO Box 19020, Curitiba, 81531-980, Paraná, Brazil
| | - Ademir Jesus Martins
- Laboratório de Fisiologia e Controle de Artrópodes Vetores, Instituto Oswaldo Cruz-FIOCRUZ, Av. Brasil 4365, Rio de Janeiro-RJ, PO Box 2104-900, Brazil. .,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil.
| | - Mário Antônio Navarro-Silva
- Laboratório de Entomologia Médica e Veterinária, Setor de Ciências Biológicas, Departamento de Zoologia, Universidade Federal do Paraná, PO Box 19020, Curitiba, 81531-980, Paraná, Brazil
| |
Collapse
|
26
|
Beesetti H, Khanna N, Swaminathan S. Investigational drugs in early development for treating dengue infection. Expert Opin Investig Drugs 2016; 25:1059-69. [DOI: 10.1080/13543784.2016.1201063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Status of vaccine research and development of vaccines for dengue. Vaccine 2016; 34:2934-2938. [DOI: 10.1016/j.vaccine.2015.12.073] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/30/2015] [Indexed: 11/21/2022]
|
28
|
McBurney SP, Sunshine JE, Gabriel S, Huynh JP, Sutton WF, Fuller DH, Haigwood NL, Messer WB. Evaluation of protection induced by a dengue virus serotype 2 envelope domain III protein scaffold/DNA vaccine in non-human primates. Vaccine 2016; 34:3500-7. [PMID: 27085173 DOI: 10.1016/j.vaccine.2016.03.108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 03/17/2016] [Accepted: 03/20/2016] [Indexed: 11/26/2022]
Abstract
We describe the preclinical development of a dengue virus vaccine targeting the dengue virus serotype 2 (DENV2) envelope domain III (EDIII). This study provides proof-of-principle that a dengue EDIII protein scaffold/DNA vaccine can protect against dengue challenge. The dengue vaccine (EDIII-E2) is composed of both a protein particle and a DNA expression plasmid delivered simultaneously via intramuscular injection (protein) and gene gun (DNA) into rhesus macaques. The protein component can contain a maximum of 60 copies of EDIII presented on a multimeric scaffold of Geobacillus stearothermophilus E2 proteins. The DNA component is composed of the EDIII portion of the envelope gene cloned into an expression plasmid. The EDIII-E2 vaccine elicited robust antibody responses to DENV2, with neutralizing antibody responses detectable following the first boost and reaching titers of greater than 1:100,000 following the second and final boost. Vaccinated and naïve groups of macaques were challenged with DENV2. All vaccinated macaques were protected from detectable viremia by infectious assay, while naïve animals had detectable viremia for 2-7 days post-challenge. All naïve macaques had detectable viral RNA from day 2-10 post-challenge. In the EDIII-E2 group, three macaques were negative for viral RNA and three were found to have detectable viral RNA post challenge. Viremia onset was delayed and the duration was shortened relative to naïve controls. The presence of viral RNA post-challenge corresponded to a 10-30-fold boost in neutralization titers 28 days post challenge, whereas no boost was observed in the fully protected animals. Based on these results, we determine that pre-challenge 50% neutralization titers of >1:6000 correlated with sterilizing protection against DENV2 challenge in EDIII-E2 vaccinated macaques. Identification of the critical correlate of protection for the EDIII-E2 platform in the robust non-human primate model lays the groundwork for further development of a tetravalent EDIII-E2 dengue vaccine.
Collapse
Affiliation(s)
- Sean P McBurney
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Justine E Sunshine
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA
| | - Sarah Gabriel
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA
| | - Jeremy P Huynh
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA
| | - William F Sutton
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Deborah H Fuller
- Department of Microbiology, University of Washington School of Medicine, 1705 NE Pacific St., Seattle, WA 98195, USA
| | - Nancy L Haigwood
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA
| | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA; Division of Infectious Diseases, Department of Medicine, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA.
| |
Collapse
|
29
|
Kirkpatrick BD, Whitehead SS, Pierce KK, Tibery CM, Grier PL, Hynes NA, Larsson CJ, Sabundayo BP, Talaat KR, Janiak A, Carmolli MP, Luke CJ, Diehl SA, Durbin AP. The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a human challenge model. Sci Transl Med 2016; 8:330ra36. [PMID: 27089205 DOI: 10.1126/scitranslmed.aaf1517] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 02/05/2016] [Indexed: 11/02/2022]
Abstract
A dengue human challenge model can be an important tool to identify candidate dengue vaccines that should be further evaluated in large efficacy trials in endemic areas. Dengue is responsible for about 390 million infections annually. Protective efficacy results for the most advanced dengue vaccine candidate (CYD) were disappointing despite its ability to induce neutralizing antibodies against all four dengue virus (DENV) serotypes. TV003 is a live attenuated tetravalent DENV vaccine currently in phase 2 evaluation. To better assess the protective efficacy of TV003, a randomized double-blind, placebo-controlled trial in which recipients of TV003 or placebo were challenged 6 months later with a DENV-2 strain, rDEN2Δ30, was conducted. The primary endpoint of the trial was protection against dengue infection, defined as rDEN2Δ30 viremia. Secondary endpoints were protection against rash and neutropenia. All 21 recipients of TV003 who were challenged with rDEN2Δ30 were protected from infection with rDEN2Δ30. None developed viremia, rash, or neutropenia after challenge. In contrast, 100% of the 20 placebo recipients who were challenged with rDEN2Δ30 developed viremia, 80% developed rash, and 20% developed neutropenia. TV003 induced complete protection against challenge with rDEN2Δ30 administered 6 months after vaccination. TV003 will be further evaluated in dengue-endemic areas. The controlled dengue human challenge model can accelerate vaccine development by evaluating the protection afforded by the vaccine, thereby eliminating poor candidates from further consideration before the initiation of large efficacy trials.
Collapse
Affiliation(s)
- Beth D Kirkpatrick
- Vaccine Testing Center, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05401, USA
| | - Stephen S Whitehead
- National Institutes of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Kristen K Pierce
- Vaccine Testing Center, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05401, USA
| | - Cecilia M Tibery
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Palmtama L Grier
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Noreen A Hynes
- Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Catherine J Larsson
- Vaccine Testing Center, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05401, USA
| | - Beulah P Sabundayo
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Anna Janiak
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Marya P Carmolli
- Vaccine Testing Center, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05401, USA
| | - Catherine J Luke
- Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sean A Diehl
- Vaccine Testing Center, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05401, USA
| | - Anna P Durbin
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
30
|
Osorio JE, Wallace D, Stinchcomb DT. A recombinant, chimeric tetravalent dengue vaccine candidate based on a dengue virus serotype 2 backbone. Expert Rev Vaccines 2016; 15:497-508. [DOI: 10.1586/14760584.2016.1128328] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
31
|
Abstract
The dengue virus and related flaviviruses are an increasing global health threat. In this perspective, we comment on and review medicinal chemistry efforts aimed at the prevention or treatment of dengue infections. We include target-based approaches aimed at viral or host factors and results from phenotypic screenings in cellular assay systems for viral replication. This perspective is limited to the discussion of results that provide explicit chemistry or structure-activity relationship (SAR), or appear to be of particular interest to the medicinal chemist for other reasons. The discovery and development efforts discussed here may at least partially be extrapolated toward other emerging flaviviral infections, such as West Nile virus. Therefore, this perspective, although not aimed at flaviviruses in general, should also be able to provide an overview of the medicinal chemistry of these closely related infectious agents.
Collapse
Affiliation(s)
- Mira A M Behnam
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University , Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Christoph Nitsche
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University , Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Veaceslav Boldescu
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University , Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.,Laboratory of Organic Synthesis, Institute of Chemistry of the Academy of Sciences of Moldova , Academiei 3, 2028 Chisinau, Moldova
| | - Christian D Klein
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University , Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Balasingam S, Horby P, Wilder-Smith A. The potential for a controlled human infection platform in Singapore. Singapore Med J 2015; 55:456-61. [PMID: 25273928 DOI: 10.11622/smedj.2014114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
For over 100 years, controlled human infection (CHI) studies have been performed to advance the understanding of the pathogenesis, treatment and prevention of infectious diseases. This methodology has seen a resurgence, as it offers an efficient model for selecting the most promising agents for further development from available candidates. CHI studies are utilised to bridge safety and immunogenicity testing and phase II/III efficacy studies. However, as this platform is not currently utilised in Asia, opportunities to study therapeutics and vaccines for infections that are important in Asia are missed. This review examines the regulatory differences for CHI studies between countries and summarises other regulatory differences in clinical trials as a whole. We found that the regulations that would apply to CHI studies in Singapore closely mirror those in the United Kingdom, and conclude that the regulatory and ethical guidelines in Singapore are compatible with the conduct of CHI studies.
Collapse
Affiliation(s)
- Shobana Balasingam
- Department of Microbiology, National University of Singapore, Centre for Life Sciences, 28 Medical Drive,Singapore 117456.
| | | | | |
Collapse
|
33
|
Larsen CP, Whitehead SS, Durbin AP. WITHDRAWN: Dengue Human Infection Models to Advance Dengue Vaccine Development. Vaccine 2015:S0264-410X(15)01424-3. [PMID: 26501186 DOI: 10.1016/j.vaccine.2015.09.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/03/2015] [Accepted: 09/28/2015] [Indexed: 11/24/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, http://dx.doi.org/10.1016/j.vaccine.2015.09.052. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Christian P Larsen
- Johns Hopkins Bloomberg School of Public Health, Department of International Health, Baltimore, MD 21205, USA
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Anna P Durbin
- Johns Hopkins Bloomberg School of Public Health, Department of International Health, Baltimore, MD 21205, USA; Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
34
|
Larsen CP, Whitehead SS, Durbin AP. Dengue human infection models to advance dengue vaccine development. Vaccine 2015; 33:7075-82. [PMID: 26424605 DOI: 10.1016/j.vaccine.2015.09.052] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
Abstract
Dengue viruses (DENV) currently infect approximately 400 million people each year causing millions to seek care and overwhelming the health care infrastructure in endemic areas. Vaccines to prevent dengue and therapeutics to treat dengue are not currently available. The efficacy of the most advanced candidate vaccine against symptomatic dengue in general and DENV-2 in particular was much lower than expected, despite the ability of the vaccine to induce neutralizing antibody against all four DENV serotypes. Because seroconversion to the DENV serotypes following vaccination was thought to be indicative of induced protection, these results have made it more difficult to assess which candidate vaccines should or should not be evaluated in large studies in endemic areas. A dengue human infection model (DHIM) could be extremely valuable to down-select candidate vaccines or therapeutics prior to engaging in efficacy trials in endemic areas. Two DHIM have been developed to assess the efficacy of live attenuated tetravalent (LATV) dengue vaccines. The first model, developed by the Laboratory of Infectious Diseases at the U. S. National Institutes of Health, utilizes a modified DENV-2 strain DEN2Δ30. This virus was derived from the DENV-2 Tonga/74 that caused only very mild clinical infection during the outbreak from which it was recovered. DEN2Δ30 induced viremia in 100%, rash in 80%, and neutropenia in 27% of the 30 subjects to whom it was given. The Walter Reed Army Institute of Research (WRAIR) is developing a DHIM the goal of which is to identify DENV that cause symptomatic dengue fever. WRAIR has evaluated seven viruses and has identified two that meet dengue fever criteria. Both of these models may be very useful in the evaluation and down-selection of candidate dengue vaccines and therapeutics.
Collapse
Affiliation(s)
- Christian P Larsen
- Johns Hopkins Bloomberg School of Public Health, Department of International Health, Baltimore, MD 21205, United States
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Anna P Durbin
- Johns Hopkins Bloomberg School of Public Health, Department of International Health, Baltimore, MD 21205, United States; Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States.
| |
Collapse
|
35
|
Animal models for studying dengue pathogenesis and therapy. Antiviral Res 2015; 123:5-14. [PMID: 26304704 DOI: 10.1016/j.antiviral.2015.08.013] [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] [Received: 06/30/2015] [Revised: 08/05/2015] [Accepted: 08/19/2015] [Indexed: 01/02/2023]
Abstract
Development of a suitable animal model for dengue virus disease is critical for understanding pathogenesis and for preclinical testing of antiviral drugs and vaccines. Many laboratory animal models of dengue virus infection have been investigated, but the challenges of recapitulating the complete disease still remain. In this review, we provide a comprehensive coverage of existing models, from man to mouse, with a specific focus on recent advances in mouse models for addressing the mechanistic aspects of severe dengue in humans. This article forms part of a symposium in Antiviral Research on flavivirus drug discovery.
Collapse
|
36
|
|
37
|
Darton TC, Blohmke CJ, Moorthy VS, Altmann DM, Hayden FG, Clutterbuck EA, Levine MM, Hill AVS, Pollard AJ. Design, recruitment, and microbiological considerations in human challenge studies. THE LANCET. INFECTIOUS DISEASES 2015; 15:840-51. [PMID: 26026195 DOI: 10.1016/s1473-3099(15)00068-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 01/13/2015] [Accepted: 01/15/2015] [Indexed: 12/26/2022]
Abstract
Since the 18th century a wealth of knowledge regarding infectious disease pathogenesis, prevention, and treatment has been accumulated from findings of infection challenges in human beings. Partly because of improvements to ethical and regulatory guidance, human challenge studies-involving the deliberate exposure of participants to infectious substances-have had a resurgence in popularity in the past few years, in particular for the assessment of vaccines. To provide an overview of the potential use of challenge models, we present historical reports and contemporary views from experts in this type of research. A range of challenge models and practical approaches to generate important data exist and are used to expedite vaccine and therapeutic development and to support public health modelling and interventions. Although human challenge studies provide a unique opportunity to address complex research questions, participant and investigator safety is paramount. To increase the collaborative effort and future success of this area of research, we recommend the development of consensus frameworks and sharing of best practices between investigators. Furthermore, standardisation of challenge procedures and regulatory guidance will help with the feasibility for using challenge models in clinical testing of new disease intervention strategies.
Collapse
Affiliation(s)
- Thomas C Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK.
| | - Vasee S Moorthy
- Department of Immunisation, Vaccines and Biologicals, WHO, Geneva, Switzerland
| | | | - Frederick G Hayden
- Department of Medicine, University of Virginia School of Medicine, Charlottesville VA, USA
| | - Elizabeth A Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adrian V S Hill
- The Jenner Institute Laboratories, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
38
|
Correlates of Protection for M Protein-Based Vaccines against Group A Streptococcus. J Immunol Res 2015; 2015:167089. [PMID: 26101780 PMCID: PMC4458553 DOI: 10.1155/2015/167089] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/28/2015] [Accepted: 05/03/2015] [Indexed: 11/17/2022] Open
Abstract
Group A streptococcus (GAS) is known to cause a broad spectrum of illness, from pharyngitis and impetigo, to autoimmune sequelae such as rheumatic heart disease, and invasive diseases. It is a significant cause of infectious disease morbidity and mortality worldwide, but no efficacious vaccine is currently available. Progress in GAS vaccine development has been hindered by a number of obstacles, including a lack of standardization in immunoassays and the need to define human correlates of protection. In this review, we have examined the current immunoassays used in both GAS and other organisms, and explored the various challenges in their implementation in order to propose potential future directions to identify a correlate of protection and facilitate the development of M protein-based vaccines, which are currently the main GAS vaccine candidates.
Collapse
|
39
|
Achee NL, Gould F, Perkins TA, Reiner RC, Morrison AC, Ritchie SA, Gubler DJ, Teyssou R, Scott TW. A critical assessment of vector control for dengue prevention. PLoS Negl Trop Dis 2015; 9:e0003655. [PMID: 25951103 PMCID: PMC4423954 DOI: 10.1371/journal.pntd.0003655] [Citation(s) in RCA: 277] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recently, the Vaccines to Vaccinate (v2V) initiative was reconfigured into the Partnership for Dengue Control (PDC), a multi-sponsored and independent initiative. This redirection is consistent with the growing consensus among the dengue-prevention community that no single intervention will be sufficient to control dengue disease. The PDC's expectation is that when an effective dengue virus (DENV) vaccine is commercially available, the public health community will continue to rely on vector control because the two strategies complement and enhance one another. Although the concept of integrated intervention for dengue prevention is gaining increasingly broader acceptance, to date, no consensus has been reached regarding the details of how and what combination of approaches can be most effectively implemented to manage disease. To fill that gap, the PDC proposed a three step process: (1) a critical assessment of current vector control tools and those under development, (2) outlining a research agenda for determining, in a definitive way, what existing tools work best, and (3) determining how to combine the best vector control options, which have systematically been defined in this process, with DENV vaccines. To address the first step, the PDC convened a meeting of international experts during November 2013 in Washington, DC, to critically assess existing vector control interventions and tools under development. This report summarizes those deliberations.
Collapse
Affiliation(s)
- Nicole L. Achee
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Fred Gould
- Department of Entomology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - T. Alex Perkins
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert C. Reiner
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health, Bloomington, Indiana, United States of America
| | - Amy C. Morrison
- Department of Entomology and Nematology, University of California, Davis, Davis, California, United States of America
- United States Naval Medical Research Unit, No. 6, Iquitos, Peru
| | - Scott A. Ritchie
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Australia
| | - Duane J. Gubler
- Emerging Infectious Diseases Program, Duke-NUS Graduate Medical School, Singapore, Singapore
- Partnership for Dengue Control, Fondation Mérieux, Lyon, France
| | - Remy Teyssou
- Partnership for Dengue Control, Fondation Mérieux, Lyon, France
| | - Thomas W. Scott
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Entomology and Nematology, University of California, Davis, Davis, California, United States of America
- Partnership for Dengue Control, Fondation Mérieux, Lyon, France
| |
Collapse
|
40
|
Abstract
Dengue viruses have spread rapidly within countries and across regions in the past few decades, resulting in an increased frequency of epidemics and severe dengue disease, hyperendemicity of multiple dengue virus serotypes in many tropical countries, and autochthonous transmission in Europe and the USA. Today, dengue is regarded as the most prevalent and rapidly spreading mosquito-borne viral disease of human beings. Importantly, the past decade has also seen an upsurge in research on dengue virology, pathogenesis, and immunology and in development of antivirals, vaccines, and new vector-control strategies that can positively impact dengue control and prevention.
Collapse
Affiliation(s)
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
41
|
Snow GE, Haaland B, Ooi EE, Gubler DJ. Review article: Research on dengue during World War II revisited. Am J Trop Med Hyg 2014; 91:1203-17. [PMID: 25311700 DOI: 10.4269/ajtmh.14-0132] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Much of the basic clinical information about dengue infection comes from experimental human studies conducted in the 1920s and 1940s. Albert Sabin's original laboratory records from one such study were bequeathed to Duane J. Gubler. These records were reviewed and 150 experiments were included in our analyses. Persons were inoculated with dengue virus 1 (DENV-1) and DENV-2. Median fever duration was shorter in primary DENV-2 infections compared with DENV-1, although maximum temperature and severity of illness were comparable. At 1.5-9 months after primary infection, 20 persons were inoculated with the heterologous serotype. Only one person inoculated with a heterologous serotype at < 8 weeks showed development of a clinical infection with a maximum temperature of 38°C, and 7 (88%) of 8 persons inoculated with a heterologous serotype at 4-9 months post-primary infection showed development of fever. On average, persons had a shorter incubation period in secondary infection compared with primary infection.
Collapse
Affiliation(s)
- Grace E Snow
- Duke University School of Medicine, Durham, North Carolina; Centre for Quantitative Medicine, Office of Clinical Sciences, Department of Statistics and Applied Probability, and Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Benjamin Haaland
- Duke University School of Medicine, Durham, North Carolina; Centre for Quantitative Medicine, Office of Clinical Sciences, Department of Statistics and Applied Probability, and Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Eng Eong Ooi
- Duke University School of Medicine, Durham, North Carolina; Centre for Quantitative Medicine, Office of Clinical Sciences, Department of Statistics and Applied Probability, and Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Duane J Gubler
- Duke University School of Medicine, Durham, North Carolina; Centre for Quantitative Medicine, Office of Clinical Sciences, Department of Statistics and Applied Probability, and Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School, Singapore
| |
Collapse
|
42
|
Sariol CA, White LJ. Utility, limitations, and future of non-human primates for dengue research and vaccine development. Front Immunol 2014; 5:452. [PMID: 25309540 PMCID: PMC4174039 DOI: 10.3389/fimmu.2014.00452] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Abstract
Dengue is considered the most important emerging, human arboviruses, with worldwide distribution in the tropics. Unfortunately, there are no licensed dengue vaccines available or specific anti-viral drugs. The development of a dengue vaccine faces unique challenges. The four serotypes co-circulate in endemic areas, and pre-existing immunity to one serotype does not protect against infection with other serotypes, and actually may enhance severity of disease. One foremost constraint to test the efficacy of a dengue vaccine is the lack of an animal model that adequately recapitulates the clinical manifestations of a dengue infection in humans. In spite of this limitation, non-human primates (NHP) are considered the best available animal model to evaluate dengue vaccine candidates due to their genetic relatedness to humans and their ability to develop a viremia upon infection and a robust immune response similar to that in humans. Therefore, most dengue vaccines candidates are tested in primates before going into clinical trials. In this article, we present a comprehensive review of published studies on dengue vaccine evaluations using the NHP model, and discuss critical parameters affecting the usefulness of the model. In the light of recent clinical data, we assess the ability of the NHP model to predict immunological parameters of vaccine performances in humans and discuss parameters that should be further examined as potential correlates of protection. Finally, we propose some guidelines toward a more standardized use of the model to maximize its usefulness and to better compare the performance of vaccine candidates from different research groups.
Collapse
Affiliation(s)
- Carlos A Sariol
- Department of Microbiology and Medical Zoology, Caribbean Primate Research Center, University of Puerto Rico-Medical Sciences Campus , San Juan, PR , USA ; Department of Internal Medicine, Caribbean Primate Research Center, University of Puerto Rico-Medical Sciences Campus , San Juan, PR , USA
| | - Laura J White
- Global Vaccine Incorporation , Research Triangle Park, NC , USA
| |
Collapse
|
43
|
Abstract
Dengue is the most common arboviral disease of humans. There is an unmet need for a therapeutic intervention that reduces the duration and severity of dengue symptoms and diminishes the likelihood of severe complications. To this end, there are active discovery efforts in industry and academia to develop interventions, with a focus on small molecule inhibitors of dengue virus replication that are suitable for therapy or chemoprophylaxis. Advancements in animal models of dengue virus infection together with the possibility of a dengue human infection model have further enhanced the platform for dengue drug discovery. Whilst drug discovery efforts gestate, there are ongoing clinical research designed to benefit today's patients, including trials of supportive care interventions, and descriptive studies that should improve the ability of clinicians to make an accurate diagnosis early in the illness course and to identify patients most at risk of progression to severe disease. This review provides a state of the art summary of dengue drug discovery, clinical trials, and supportive allied research and reflects discussions at the 2nd International Dengue Therapeutics Workshop held in Ho Chi Minh City, Vietnam, in December 2013.
Collapse
|
44
|
Abstract
Dengue virus (DENV) is a significant cause of morbidity and mortality in tropical and subtropical regions, causing hundreds of millions of infections each year. Infections range from asymptomatic to a self-limited febrile illness, dengue fever (DF), to the life-threatening dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). The expanding of the habitat of DENV-transmitting mosquitoes has resulted in dramatic increases in the number of cases over the past 50 years, and recent outbreaks have occurred in the United States. Developing a dengue vaccine is a global health priority. DENV vaccine development is challenging due to the existence of four serotypes of the virus (DENV1-4), which a vaccine must protect against. Additionally, the adaptive immune response to DENV may be both protective and pathogenic upon subsequent infection, and the precise features of protective versus pathogenic immune responses to DENV are unknown, complicating vaccine development. Numerous vaccine candidates, including live attenuated, inactivated, recombinant subunit, DNA, and viral vectored vaccines, are in various stages of clinical development, from preclinical to phase 3. This review will discuss the adaptive immune response to DENV, dengue vaccine challenges, animal models used to test dengue vaccine candidates, and historical and current dengue vaccine approaches.
Collapse
Affiliation(s)
- Lauren E Yauch
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Sujan Shresta
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA.
| |
Collapse
|
45
|
Abstract
The average effect of an infectious disease intervention (eg, a vaccine) varies across populations with different degrees of exposure to the pathogen. As a result, many investigators favor a per-exposure effect measure that is considered independent of the population level of exposure and that can be used in simulations to estimate the total disease burden averted by an intervention across different populations. However, while per-exposure effects are frequently estimated, the quantity of interest is often poorly defined, and assumptions in its calculation are typically left implicit. In this article, we build upon work by Halloran and Struchiner (Epidemiology. 1995;6:142-151) to develop a formal definition of the per-exposure effect and discuss conditions necessary for its unbiased estimation. With greater care paid to the parameterization of transmission models, their results can be better understood and can thereby be of greater value to decision-makers.
Collapse
|
46
|
Rothman AL. DHIM supporting immunologic investigations and the identification of immune correlates of protection. J Infect Dis 2014; 209 Suppl 2:S61-5. [PMID: 24872398 DOI: 10.1093/infdis/jiu111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Evidence suggesting that immune responses to dengue virus (DENV) have the potential for both beneficial and detrimental effects on the outcome of infection is a concern for dengue vaccine development. There is thus a great need to define measures of DENV-specific immune responses that reliably indicate when immunity is protective. The existence of 4 main DENV serotypes and the difficulty in defining which individuals have been exposed and to which viruses present challenges to defining immune correlates of protective immunity against DENV in field efficacy studies; experimental infection studies in humans offer a pathway to address these challenges.
Collapse
Affiliation(s)
- Alan L Rothman
- Institute for Immunology and Informatics, College of the Environment and Life Sciences, University of Rhode Island, Providence
| |
Collapse
|
47
|
Mores CN, Christofferson RC, Davidson SA. The role of the mosquito in a dengue human infection model. J Infect Dis 2014; 209 Suppl 2:S71-8. [PMID: 24872400 DOI: 10.1093/infdis/jiu110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent efforts to combat the growing global threat of dengue disease, including deployment of phase IIb vaccine trials, has continued to be hindered by uncertainty surrounding equitable immune responses of serotypes, relative viral fitness of vaccine vs naturally occurring strains, and the importance of altered immune environments due to natural delivery routes. Human infection models can significantly improve our understanding of the importance of certain phenotypic characteristics of viral strains, and inform strain selection and trial design. With human models, we can further assess the importance of the natural delivery route of DENV and/or the accompanying mosquito salivary milieu. Accordingly, we discuss the use of mosquitoes in such a human infection model with DENV, identify important considerations, and make preliminary recommendations for deployment of such a mosquito improved DENV human infection model (miDHIM).
Collapse
Affiliation(s)
- Christopher N Mores
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rebecca C Christofferson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Silas A Davidson
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, Maryland
| |
Collapse
|
48
|
Abstract
Dengue is a global health problem and of concern to travelers and deploying military personnel with development and licensure of an effective tetravalent dengue vaccine a public health priority. The dengue viruses (DENVs) are mosquito-borne flaviviruses transmitted by infected Aedes mosquitoes. Illness manifests across a clinical spectrum with severe disease characterized by intravascular volume depletion and hemorrhage. DENV illness results from a complex interaction of viral properties and host immune responses. Dengue vaccine development efforts are challenged by immunologic complexity, lack of an adequate animal model of disease, absence of an immune correlate of protection, and only partially informative immunogenicity assays. A dengue human infection model (DHIM) will be an essential tool in developing potential dengue vaccines or antivirals. The potential performance parameters needed for a DHIM to support vaccine or antiviral candidates are discussed.
Collapse
Affiliation(s)
- Timothy P Endy
- Infectious Disease Division, Department of Medicine, State University of New York, Upstate Medical University, Syracuse
| |
Collapse
|
49
|
Lyons AG. The human dengue challenge experience at the Walter Reed Army Institute of Research. J Infect Dis 2014; 209 Suppl 2:S49-55. [PMID: 24872396 DOI: 10.1093/infdis/jiu174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent discordance between measured levels of serotypes of dengue virus neutralizing antibody and clinical outcomes suggests a need to reevaluate the process of prescreening dengue vaccine candidates to better predict their clinical benefit before initiation of large-scale human vaccine trials. In the absence of a reliable animal model for dengue, a human dengue virus challenge model (ie, a controlled live dengue virus infectious challenge study) may prove useful and timely to elucidate mechanisms that underlie protection (as well as virulence), thus facilitating down-selection of vaccine candidates before beginning advanced field trials. Dengue challenge studies were safely used in prior decades to study the vector biology, clinical spectrum of illness, and reactogenicity of candidate live dengue virus vaccines of uncertain attenuation. Redeveloping the human dengue challenge model following current regulatory guidance, good manufacturing practice, and good clinical practice could streamline and accelerate vaccine development by offering a time- and resource-efficient method to evaluate the safety and potential efficacy of dengue vaccine and therapeutic candidates. In this article, the development of such a challenge model and its subsequent application is summarized from 2 recent reports.
Collapse
|
50
|
Abstract
Dengue is a arboviral infection that represents a major global health burden. There is an unmet need for effective dengue therapeutics to reduce symptoms, duration of illness and incidence of severe complications. Here, we consider the merits of a dengue human infection model (DHIM) for drug development. A DHIM could allow experimentally controlled studies of candidate therapeutics in preselected susceptible volunteers, potentially using smaller sample sizes than trials that recruited patients with dengue in an endemic country. In addition, the DHIM would assist the conduct of intensive pharmacokinetic and basic research investigations and aid in determining optimal drug dosage. Furthermore, a DHIM could help establish proof of concept that chemoprophylaxis against dengue is feasible. The key challenge in developing the DHIM for drug development is to ensure the model reliably replicates the typical clinical and laboratory features of naturally acquired, symptomatic dengue.
Collapse
Affiliation(s)
- James Whitehorn
- Department of Clinical Research, London School of Hygiene and Tropical Medicine
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases
| | | | - Cameron P. Simmons
- Centre for Tropical Medicine, Oxford University, United Kingdom
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases
- Nossal Institute for Global Health and Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|