1
|
de Almeida CMF, Rios WM, Duarte MPF, Brandão IT, Paiva NF, Vicentini FTMDC. Self-assembled nanovaccine based on apoferritin: Development and vaccine regimen evaluation. Eur J Pharm Biopharm 2025; 206:114589. [PMID: 39613269 DOI: 10.1016/j.ejpb.2024.114589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/18/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Apoferritin-based systems have been explored last decade for their potential as vaccine delivery for viral diseases. The nanosized properties of an apoferritin-based system could increase immunogenicity, contribute to antigen stability, and reduce the vaccines' adverse effects. The mutated extracellular portion of the epidermal growth factor receptor (EGFRvIII peptide, PEPvIII) can be applied as a specific tumoral antigen due to rare expression in normal cells. In this context, the present study proposed the development and the immunogenicity evaluation of an apoferritin-based system (AFt) to deliver a peptide vaccine for an antitumoral purpose. We developed a formulation with different PEPvIII:AFt ratios and during the association efficiency analysis, identified the dependence between the AFt concentration and the PEPvIII association percentage in the formulation. Besides, differences in the immune responses against EGFRvIII were observed depending on the PEPvIII concentration due to the different association efficiencies. Finally, the humoral immune response results showed a high antibody production against AFt, which might affect the immunological tolerance. Collectively, this study establishes the PEPvIII:AFt formulation process and highlights the determinant factors for guaranteeing vaccine safety and efficacy.
Collapse
Affiliation(s)
| | - Wendy Martin Rios
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | | | - Izaíra Tincani Brandão
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | - Natalia Floriano Paiva
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | | |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 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
|
3
|
Singh RK, Tiwari A, Satone PD, Priya T, Meshram RJ. Updates in the Management of Dengue Shock Syndrome: A Comprehensive Review. Cureus 2023; 15:e46713. [PMID: 38021722 PMCID: PMC10631559 DOI: 10.7759/cureus.46713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/08/2023] [Indexed: 12/01/2023] Open
Abstract
Dengue is a very serious public health problem that can manifest a wide range of symptoms from asymptomatic to fatal conditions, such as dengue shock syndrome (DSS). It is a life-threatening mosquito-borne viral infection widely spread in tropical areas. Dengue virus transmission occurs from an infected Aedes mosquito to humans. Various factors are responsible for the occurrence of the disease, such as viral load, age of the host, immune status of the host, and genetic variability. Dengue infection occurs in three phases: febrile, critical, and recovery. The febrile phase lasts for seven days and manifests symptoms such as high-grade fever, headache, arthralgia, and backache, and in some cases, the upper respiratory tract and gastrointestinal tract are also involved. Severe dengue is characterized by endothelial dysfunction that causes vascular permeability and plasma leakage. The fundamental mechanisms of these immune pathologies are not yet known. Dengue manifests various complications such as dengue encephalopathy, encephalitis, stroke, ocular involvement, acute transverse myelitis, myalgia, and cerebellar syndrome, but the most commonly seen is liver involvement. Dengue is managed supportively because there are no proven curative treatments. The cornerstone of care during the critical period of dengue is prudent fluid resuscitation. The first fluid of preference is a crystalloid. Prophylactic transfusion of platelets is not advised. The occurrence of four antigenically different dengue virus serotypes, each able to elicit a cross-reactive and disease-enhancing antibody response against the other three serotypes, has made the creation of the dengue vaccine a difficult undertaking. The development of a dengue vaccine has faced significant challenges due to a lack of the best animal models and a variety of immunological conditions in people, particularly in endemic locations. Dengvaxia is a live attenuated vaccine, which was developed by Sanofi. It is made up of four chimeric vaccine viruses produced by Vero cells.
Collapse
Affiliation(s)
- Rakshit K Singh
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aakriti Tiwari
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prasiddhi D Satone
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Tannu Priya
- Department of Paediatrics, Pravara Institute of Medical Sciences, Shirdi, IND
| | - Revat J Meshram
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
4
|
Malik S, Ahsan O, Mumtaz H, Tahir Khan M, Sah R, Waheed Y. Tracing down the Updates on Dengue Virus-Molecular Biology, Antivirals, and Vaccine Strategies. Vaccines (Basel) 2023; 11:1328. [PMID: 37631896 PMCID: PMC10458802 DOI: 10.3390/vaccines11081328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Nearly half of the world is at risk of developing dengue infection. Dengue virus is the causative agent behind this public healthcare concern. Millions of dengue cases are reported every year, leading to thousands of deaths. The scientific community is working to develop effective therapeutic strategies in the form of vaccines and antiviral drugs against dengue. METHODS In this review, a methodological approach has been used to gather data from the past five years to include the latest developments against the dengue virus. RESULTS Different therapeutics and antiviral targets against the dengue virus are at different stages of development, but none have been approved by the FDA. Moreover, various vaccination strategies have also been discussed, including attenuated virus vaccines, recombinant subunit vaccines, viral vector vaccines, DNA vaccines, nanotechnology, and plant-based vaccines, which are used to develop effective vaccines for the dengue virus. Many dengue vaccines pass the initial phases of evaluation, but only two vaccines have been approved for public use. DENGVAXIA is the only FDA-approved vaccine against all four stereotypes of the dengue virus, but it is licensed for use only in individuals 6-16 years of age with laboratory-confirmed previous dengue infection and living in endemic countries. Takeda is the second vaccine approved for use in the European Union, the United Kingdom, Brazil, Argentina, Indonesia, and Thailand. It produced sustained antibody responses against all four serotypes of dengue virus, regardless of previous exposure and dosing schedule. Other dengue vaccine candidates at different stages of development are TV-003/005, TDENV PIV, V180, and some DNA vaccines. CONCLUSION There is a need to put more effort into developing effective vaccines and therapeutics for dengue, as already approved vaccines and therapeutics have limitations. DENGVAXIA is approved for use in children and teenagers who are 6-16 years of age and have confirmed dengue infection, while Takeda is approved for use in certain countries, and it has withdrawn its application for FDA approval.
Collapse
Affiliation(s)
- Shiza Malik
- Bridging Health Foundation, Rawalpindi 46000, Pakistan
| | - Omar Ahsan
- Department of Medicine, Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan
| | - Hassan Mumtaz
- Innovation, Implementation, and Partnership Unit, Association for Social Development, Islamabad 44000, Pakistan
- Health Services Academy, Islamabad 44000, Pakistan
| | - Muhammad Tahir Khan
- Institute of Molecular Biology and Biotechnology (IMBB), University of Lahore, 1KM Defence Road, Lahore 58810, Pakistan
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang 473006, China
| | - Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu 44600, Nepal
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
| | - Yasir Waheed
- Office of Research, Innovation and Commercialization (ORIC), Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad 44000, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos 1401, Lebanon
| |
Collapse
|
5
|
Cohen G, Jungsomsri P, Sangwongwanich J, Tawinprai K, Siripongboonsitti T, Porntharukchareon T, Wittayasak K, Thonwirak N, Soonklang K, Sornsamdang G, Auewarakul C, Mahanonda N. Immunogenicity and reactogenicity after heterologous prime-boost vaccination with CoronaVac and ChAdox1 nCov-19 (AZD1222) vaccines. Hum Vaccin Immunother 2022; 18:2052525. [PMID: 35323079 PMCID: PMC9115782 DOI: 10.1080/21645515.2022.2052525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mass vaccination with a safe and effective vaccine may be the best way to control the COVID-19 pandemic. Heterologous prime-boost vaccination with the CoronaVac and AZD1222 vaccines may increase the immunogenicity elicited by either vaccine alone. This study sought to compare the immunogenicity of a heterologous CoronaVac and AZD1222 prime-boost with a homologous CoronaVac prime-boost. From July 13 to September 2, 2021, 88 participants were enrolled in the study. Half (n = 44) of the participants were assigned to the AZD1222/CoronaVac cohort and half were assigned to the CoronaVac/AZD1222 cohort. Both cohorts had a prime-boost interval of 4 weeks. A control group of 136 health care personnel who received the homologous CoronaVac/CoronaVac prime-boost was matched by age and sex to the experimental cohorts. The primary endpoint was the geometric mean ratio (GMR) of the anti-receptor binding domain (RBD) antibody concentration 4 weeks after the booster dose was administered. The CoronaVac/CoronaVac cohort served as the reference group. Baseline age and sex were similar, and the median age was 42.5 years. The GMR was 2.58 (95% confidence interval [CI] 1.80–3.71) and 8.69 (95% CI 6.05–12.47) in the AZD1222/CoronaVac and CoronaVac/AZD1222 cohorts, respectively. Reactogenicity was similar following prime and booster doses with the same vaccine. Findings indicated that the heterologous CoronaVac and AZD1222 prime-boost combination elicited a more robust immune response than the homologous CoronaVac prime-boost. While both heterologous prime-boost combinations showed similar reactogenicity, the immunogenicity of the CoronaVac/AZD1222 cohort was higher, indicating that the order of prime-boost vaccine administration was important.
Collapse
Affiliation(s)
- Guy Cohen
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Pawornrath Jungsomsri
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Jirath Sangwongwanich
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Kriangkrai Tawinprai
- Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | | | - Kasiruck Wittayasak
- Center of Learning and Research in Celebration of HRH Princess Chulabhorn's 60th Birthday Anniversary, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Nawarat Thonwirak
- Center of Learning and Research in Celebration of HRH Princess Chulabhorn's 60th Birthday Anniversary, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Kamonwan Soonklang
- Center of Learning and Research in Celebration of HRH Princess Chulabhorn's 60th Birthday Anniversary, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Gaidganok Sornsamdang
- Central Laboratory Center, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Chirayu Auewarakul
- Center of Learning and Research in Celebration of HRH Princess Chulabhorn's 60th Birthday Anniversary, Chulabhorn Royal Academy, Bangkok, Thailand.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Nithi Mahanonda
- Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
6
|
Odio CD, Katzelnick LC. 'Mix and Match' vaccination: Is dengue next? Vaccine 2022; 40:6455-6462. [PMID: 36195473 PMCID: PMC9526515 DOI: 10.1016/j.vaccine.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 01/27/2023]
Abstract
The severity of the COVID-19 pandemic and the development of multiple SARS-CoV-2 vaccines expedited vaccine 'mix and match' trials in humans and demonstrated the benefits of mixing vaccines that vary in formulation, strength, and immunogenicity. Heterologous sequential vaccination may be an effective approach for protecting against dengue, as this strategy would mimic the natural route to broad dengue protection and may overcome the imbalances in efficacy of the individual leading live attenuated dengue vaccines. Here we review 'mix and match' vaccination trials against SARS-CoV-2, HIV, and dengue virus and discuss the possible advantages and concerns of future heterologous immunization with the leading dengue vaccines. COVID-19 trials suggest that priming with a vaccine that induces strong cellular responses, such as an adenoviral vectored product, followed by heterologous boost may optimize T cell immunity. Moreover, heterologous vaccination may induce superior humoral immunity compared to homologous vaccination when the priming vaccine induces a narrower response than the boost. The HIV trials reported that heterologous vaccination was associated with broadened antigen responses and that the sequence of the vaccines significantly impacts the regimen's immunogenicity and efficacy. In heterologous dengue immunization trials, where at least one dose was with a live attenuated vaccine, all reported equivalent or increased immunogenicity compared to homologous boost, although one study reported increased reactogenicity. The three leading dengue vaccines have been evaluated for safety and efficacy in thousands of study participants but not in combination in heterologous dengue vaccine trials. Various heterologous regimens including different combinations and sequences should be trialed to optimize cellular and humoral immunity and the breadth of the response while limiting reactogenicity. A blossoming field dedicated to more accurate correlates of protection and enhancement will help confirm the safety and efficacy of these strategies.
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, United States
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, United States.
| |
Collapse
|
7
|
DÜZENLİ ÖF, OKAY S, KAZKAYASI İ, ÖNER AF. Recombinant AhpC antigen from Mycobacterium bovis boosts BCG-primed immunity in mice. Turk J Biol 2021; 46:95-104. [PMID: 37533671 PMCID: PMC10393101 DOI: 10.3906/biy-2108-41] [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: 08/25/2021] [Revised: 02/08/2022] [Accepted: 11/14/2021] [Indexed: 02/01/2023] Open
Abstract
Tuberculosis (TB) is still one of the most common infectious diseases around the world despite the widespread use of BCG (bacille Calmette-Guerin) strain of Mycobacterium bovis as a vaccine. This vaccine does not always protect people from TB, and, thus, new effective vaccines or vaccination strategies are being investigated. In this study, alkyl hydroperoxide reductase (AhpC) from M. bovis was evaluated as a new candidate vaccine antigen against TB in BALB/c mice model. The ahpC gene was amplified from M.bovis genome, cloned, and expressed in Escherichia coli. Vaccine antigen AhpC was formulated with Montanide ISA 61 VG, an oil-based emulsion adjuvant. Both IgG and IL-12 responses were observed in mice after administering the formulation both as a subunit vaccine alone and also as a booster vaccine for BCG immunization. However, a long-lasting response was observed when AhpC formulation was used as a booster (for BCG-primed immunization) as compared to being used as a subunit vaccine alone. In short, these findings suggested that AhpC has the potential to be used as a booster vaccine candidate for BCG-primed immunization.
Collapse
Affiliation(s)
- Özgün Fırat DÜZENLİ
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Hacettepe University, Ankara,
Turkey
| | - Sezer OKAY
- Department of Vaccine Technology, Vaccine Institute, Hacettepe University, Ankara,
Turkey
| | - İnci KAZKAYASI
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University, Ankara,
Turkey
| | - Ayşe Filiz ÖNER
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Hacettepe University, Ankara,
Turkey
| |
Collapse
|
8
|
Stress-Induced Epstein-Barr Virus Reactivation. Biomolecules 2021; 11:biom11091380. [PMID: 34572593 PMCID: PMC8470332 DOI: 10.3390/biom11091380] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Epstein-Barr virus (EBV) is typically found in a latent, asymptomatic state in immunocompetent individuals. Perturbations of the host immune system can stimulate viral reactivation. Furthermore, there are a myriad of EBV-associated illnesses including various cancers, post-transplant lymphoproliferative disease, and autoimmune conditions. A thorough understanding of this virus, and the interplay between stress and the immune system, is essential to establish effective treatment. This review will provide a summary of the interaction between both psychological and cellular stressors resulting in EBV reactivation. It will examine mechanisms by which EBV establishes and maintains latency and will conclude with a brief overview of treatments targeting EBV.
Collapse
|
9
|
Enhanced dengue vaccine virus replication and neutralizing antibody responses in immune primed rhesus macaques. NPJ Vaccines 2021; 6:77. [PMID: 34021159 PMCID: PMC8140083 DOI: 10.1038/s41541-021-00339-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
Antibody-dependent enhancement (ADE) is suspected to influence dengue virus (DENV) infection, but the role ADE plays in vaccination strategies incorporating live attenuated virus components is less clear. Using a heterologous prime-boost strategy in rhesus macaques, we examine the effect of priming with DENV purified inactivated vaccines (PIVs) on a tetravalent live attenuated vaccine (LAV). Sera exhibited low-level neutralizing antibodies (NAb) post PIV priming, yet moderate to high in vitro ADE activity. Following LAV administration, the PIV primed groups exhibited DENV-2 LAV peak viremias up to 1,176-fold higher than the mock primed group, and peak viremia correlated with in vitro ADE. Furthermore, PIV primed groups had more balanced and higher DENV-1–4 NAb seroconversion and titers than the mock primed group following LAV administration. These results have implications for the development of effective DENV vaccine prime-boost strategies and for our understanding of the role played by ADE in modulating DENV replication.
Collapse
|
10
|
Alves AMB, Costa SM, Pinto PBA. Dengue Virus and Vaccines: How Can DNA Immunization Contribute to This Challenge? FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:640964. [PMID: 35047911 PMCID: PMC8757892 DOI: 10.3389/fmedt.2021.640964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/17/2021] [Indexed: 01/02/2023] Open
Abstract
Dengue infections still have a tremendous impact on public health systems in most countries in tropical and subtropical regions. The disease is systemic and dynamic with broad range of manifestations, varying from mild symptoms to severe dengue (Dengue Hemorrhagic Fever and Dengue Shock Syndrome). The only licensed tetravalent dengue vaccine, Dengvaxia, is a chimeric yellow fever virus with prM and E genes from the different dengue serotypes. However, recent results indicated that seronegative individuals became more susceptible to develop severe dengue when infected after vaccination, and now WHO recommends vaccination only to dengue seropositive people. One possibility to explain these data is the lack of robust T-cell responses and antibody-dependent enhancement of virus replication in vaccinated people. On the other hand, DNA vaccines are excellent inducers of T-cell responses in experimental animals and it can also elicit antibody production. Clinical trials with DNA vaccines have improved and shown promising results regarding the use of this approach for human vaccination. Therefore, in this paper we review preclinical and clinical tests with DNA vaccines against the dengue virus. Most of the studies are based on the E protein since this antigen is the main target for neutralizing antibody production. Yet, there are other reports with DNA vaccines based on non-structural dengue proteins with protective results, as well. Combining structural and non-structural genes may be a solution for inducing immune responses aging in different infection moments. Furthermore, DNA immunizations are also a very good approach in combining strategies for vaccines against dengue, in heterologous prime/boost regimen or even administering different vaccines at the same time, in order to induce efficient humoral and cellular immune responses.
Collapse
Affiliation(s)
- Ada Maria Barcelos Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | | |
Collapse
|
11
|
Muhialdin BJ, Zawawi N, Abdull Razis AF, Bakar J, Zarei M. Antiviral activity of fermented foods and their probiotics bacteria towards respiratory and alimentary tracts viruses. Food Control 2021; 127:108140. [PMID: 33867696 PMCID: PMC8036130 DOI: 10.1016/j.foodcont.2021.108140] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/09/2021] [Accepted: 04/04/2021] [Indexed: 02/06/2023]
Abstract
The recent COVID-19, a viral outbreak calls for a high demand for non-conventional antiviral agents that can reduce the risk of infections and promote fast recovery. Fermented foods and their probiotics bacteria have recently received increasing interest due to the reported potential of high antiviral activity. Several probiotics strains demonstrated broad range of antiviral activities and different mechanisms of action. This article will review the diversity, health benefits, interaction with immune system and antiviral activity of fermented foods and their probiotics bacteria. In addition, the mechanisms of action will be reviewed to determine the broad range potential antiviral activity against the respiratory and alimentary tracts viruses. The probiotics bacteria and bioactive compounds in fermented foods demonstrated antiviral activities against respiratory and alimentary tracts viruses. The mechanism of action was reported to be due to the stimulation of the immune system function via enhancing natural killers cell toxicity, enhance the production of pro-inflammatory cytokines, and increasing the cytotoxic of T lymphocytes (CD3+, CD16+, CD56+). However, further studies are highly recommended to determine the potential antiviral activity for traditional fermented foods.
Collapse
Affiliation(s)
- Belal J Muhialdin
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia.,Halal Products Research Institute, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia
| | - Norhasnida Zawawi
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia.,Natural Medicines and Product Research Laboratory, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia.,Natural Medicines and Product Research Laboratory, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia
| | - Jamilah Bakar
- Department of Food Technology, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM, Selangor, Malaysia
| | - Mohammad Zarei
- Department of Food Science and Technology, School of Industrial Technology, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, 40450, Selangor, Malaysia
| |
Collapse
|
12
|
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
|
13
|
Coronel-MartÍnez DL, Park J, López-Medina E, Capeding MR, Cadena Bonfanti AA, Montalbán MC, Ramírez I, Gonzales MLA, DiazGranados CA, Zambrano B, Dayan G, Savarino S, Chen Z, Wang H, Sun S, Bonaparte M, Rojas A, Ramírez JC, Verdan MA, Noriega F. Immunogenicity and safety of simplified vaccination schedules for the CYD-TDV dengue vaccine in healthy individuals aged 9-50 years (CYD65): a randomised, controlled, phase 2, non-inferiority study. THE LANCET. INFECTIOUS DISEASES 2020; 21:517-528. [PMID: 33212067 DOI: 10.1016/s1473-3099(20)30767-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Three doses of the licensed tetravalent dengue vaccine CYD-TDV (Dengvaxia, Sanofi Pasteur, Lyon France) are immunogenic and effective against symptomatic dengue in individuals aged 9 years and older who are dengue seropositive. Previous trials have provided some evidence that antibody responses elicited after just one dose or two doses of CYD-TDV might be similar to those elicited after three doses. We compared antibody responses following one-dose, two-dose, and three-dose vaccination regimens in individuals who were dengue seropositive at baseline up to 1 year after the last injection. METHODS In this randomised, controlled, phase 2, non-inferiority study (CYD65), healthy individuals aged 9-50 years were recruited from the community in three sites in Colombia and three sites in the Philippines. Participants were randomly assigned (1:1:1), using a permuted block method with stratification by site and age group, to receive, at 6-month intervals (on day 0, month 6, and month 12), three doses of CYD-TDV (three-dose group), one dose of placebo (on day 0) and two doses of CYD-TDV (at months 6 and 12; two-dose group), or two doses of placebo (on day 0 and month 6) and one dose of CYD-TDV (at month 12; one-dose group). Each dose of CYD-TDV was 0·5 mL, administered subcutaneously into the deltoid of the upper arm. Participants, study staff, investigators, and the funder were masked to group assignment. The co-primary endpoints were geometric mean titres (GMTs) of neutralising antibodies against each dengue virus serotype at 28 days and 1 year after the last vaccine injection. After a protocol amendment during the conduct of the study, the original co-primary objectives of non-inferiority of the one-dose and two-dose groups to the three-dose group were altered to include non-inferiority of the two-dose group to the three-dose group only, to be assessed in individuals who were dengue seropositive at baseline. Non-inferiority was shown if the lower limit of the 95% CI for the ratio of GMTs (GMR) at 28 days and 1 year between groups was more than 0·5 for each serotype. The analysis of the coprimary objectives was done in the per-protocol analysis dataset, which included all participants who had been vaccinated, had no protocol deviations, and had a valid serology test result for at least one dengue serotype at 28 days after the third injection. Safety was assessed throughout in all participants who received at least one injection of study drug, regardless of serostatus. This trial is registered with ClinicalTrials.gov, NCT02628444, and is closed to accrual. FINDINGS Between May 2, 2016, and Sept 16, 2016, we recruited and enrolled 1050 individuals, of whom 1048 received at least one injection and 993 had at least one blood sample taken (full-analysis dataset; 333 in three-dose group, 328 in two-dose group, and 332 in one-dose group). 860 (86·6%) of 993 participants in the full-analysis dataset were dengue seropositive at baseline. Non-inferiority (two dose vs three dose) was shown for each serotype at both 28 days and 1 year among dengue-seropositive participants (number of participants assessed: 272 [two-dose group], 265 [three-dose group] at 28 days; and 190 [two-dose group], 185 [three-dose group] at 1 year). At 28 days after the last injection, neutralising antibody GMTs were 899 (95% CI 752-1075) in the two-dose group versus 822 (700-964) in the three dose group against dengue serotype 1 (GMR 1·09 [95% CI 0·86-1·39]); 869 (754-1002) versus 875 (770-995) against serotype 2 (GMR 0·99 [0·82-1·20]); 599 (524-685) versus 610 (535-694) against serotype 3 (GMR 0·98 [0·82-1·18]); and 510 (453-575) versus 531 (470-601) against serotype 4 (GMR 0·96 [0·81-1·14]). At year 1, GMTs had decreased but remained above baseline for all serotypes: 504 (95% CI 403-630) in the two-dose group versus 490 (398-604) in the three-dose group against serotype 1 (GMR 1·03 [0·76-1·40]); 737 (611-888) versus 821 (704-957) against serotype 2 (GMR 0·90 [0·71-1·14]); 437 (368-519) versus 477 (405-561) against serotype 3 (GMR 0·92 [0·72-1·16]); and 238 (205-277) versus 270 (235-310) against serotype 4 (GMR 0·88 [0·72-1·09]). Reactogenicity profiles were similar across treatment groups. Most unsolicited adverse events after any injection were non-serious and systemic in nature. During the study, 60 serious adverse events were reported in 58 participants (14 in three-dose group, 26 in two-dose group, 18 in one-dose group), mostly infection and infestations or injury, poisoning, and procedural complications. No serious adverse events of special interest or admissions to hospital for dengue occurred. Two deaths occurred, unrelated to study treatment. INTERPRETATION A two-dose CYD-TDV regimen might be an alternative to the licensed three-dose regimen in individuals who are dengue seropositive at baseline and aged 9 years and older. Vaccination with a reduced number of doses could lead to improved vaccine compliance and coverage, especially in low-resource settings. FUNDING Sanofi Pasteur.
Collapse
Affiliation(s)
| | | | - Eduardo López-Medina
- Centro de Estudios en Infectología Pediátrica, Universidad del Valle and Centro Médico Imbanaco, Cali, Valle del Cauca, Colombia
| | - María Rosario Capeding
- Research Institute for Tropical Medicine, Filinvest Corporate City, Alabang, Muntinlupa, Metro Manila, Philippines
| | | | | | - Isabel Ramírez
- Infectious Diseases, Internal Medicine, Hospital Pablo Tobón Uribe, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | | | | | | | | | | | | | - Hao Wang
- Sanofi Pasteur, Chaoyang, Beijing, China
| | - Sunny Sun
- Sanofi Pasteur, Chaoyang, Beijing, China
| | | | | | | | | | | |
Collapse
|
14
|
Immune Response Persistence and Safety of a Booster Dose of the Tetravalent Dengue Vaccine in Adolescents and Adults Who Previously Completed the 3-dose Schedule 4-5 Years Earlier in Latin America: A Randomized Placebo-controlled Trial. Pediatr Infect Dis J 2020; 39:961-968. [PMID: 32932330 DOI: 10.1097/inf.0000000000002830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND We previously described an increased immune response 28 days after a booster dose of the live, attenuated, tetravalent dengue vaccine (CYD-TDV) in healthy adolescents and adults in Latin America (CYD64, NCT02623725). This follow-up study evaluated immune response persistence and safety of a CYD-TDV booster dose up to Month (M) 24 post-booster. METHODS This study included 250 participants who previously received 3 primary doses of CYD-TDV in the CYD13 (NCT00993447) and CYD30 (NCT01187433) studies, and who were randomized 4-5 years later to receive a CYD-TDV booster or placebo (3:1). Dengue neutralizing antibodies against the parental dengue virus strains were assessed using the plaque reduction neutralization test (PRNT50) at M6, M12, and M24 post-booster. Post-booster memory B-cell responses were assessed in a subset of participants using the FluoroSpot assay up to M12 post-booster. RESULTS In the CYD-TDV group (n = 187), dengue neutralizing antibody geometric mean titers (GMTs) declined from the peak at day 28 through to M24 for all serotypes. GMTs at M24 were similar to those at pre-booster among baseline dengue seropositives. A similar trend was observed for baseline dengue seronegatives, albeit at a lower magnitude. Previous vaccination-induced detectable B-cell memory responses in seropositives and seronegatives that decreased to pre-booster levels at M12 post-booster. The CYD-TDV booster dose was well-tolerated. CONCLUSIONS In baseline dengue seropositives, following a CYD-TDV booster dose administered 4-5 years after primary immunization, dengue neutralizing antibody GMTs and B-cell memory responses peaked in the short-term before gradually decreasing over time. A CYD-TDV booster dose could improve protection against dengue during outbreak periods.
Collapse
|
15
|
Lin L, Koren MA, Paolino KM, Eckels KH, De La Barrera R, Friberg H, Currier JR, Gromowski GD, Aronson NE, Keiser PB, Sklar MJ, Sondergaard EL, Jasper LE, Endy TP, Jarman RG, Thomas SJ. Immunogenicity of a Live-Attenuated Dengue Vaccine Using a Heterologous Prime-Boost Strategy in a Phase 1 Randomized Clinical Trial. J Infect Dis 2020; 223:1707-1716. [PMID: 32966573 DOI: 10.1093/infdis/jiaa603] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/18/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Dengue is a global health problem and the development of a tetravalent dengue vaccine with durable protection is a high priority. A heterologous prime-boost strategy has the advantage of eliciting immune responses through different mechanisms and therefore may be superior to homologous prime-boost strategies for generating durable tetravalent immunity. METHODS In this phase 1 first-in-human heterologous prime-boost study, 80 volunteers were assigned to 4 groups and received a tetravalent dengue virus (DENV-1-4) purified inactivated vaccine (TDENV-PIV) with alum adjuvant and a tetravalent dengue virus (DENV-1-4) live attenuated vaccine (TDENV-LAV) in different orders and dosing schedules (28 or 180 days apart). RESULTS All vaccination regimens had acceptable safety profiles and there were no vaccine-related serious adverse events. TDEN-PIV followed by TDEN-LAV induced higher neutralizing antibody titers and a higher rate of tetravalent seroconversions compared to TDEN-LAV followed by TDEN-PIV. Both TDEN-PIV followed by TDEN-LAV groups demonstrated 100% tetravalent seroconversion 28 days following the booster dose, which was maintained for most of these subjects through the day 180 measurement. CONCLUSIONS A heterologous prime-boost vaccination strategy for dengue merits additional evaluation for safety, immunogenicity, and potential for clinical benefit. CLINICAL TRIALS REGISTRATION NCT02239614.
Collapse
Affiliation(s)
- Leyi Lin
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Michael A Koren
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Kristopher M Paolino
- Clinical Trials Center, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Kenneth H Eckels
- Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Rafael De La Barrera
- Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Heather Friberg
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Gregory D Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Naomi E Aronson
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Paul B Keiser
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Marvin J Sklar
- Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Erica L Sondergaard
- Clinical Trials Center, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Louis E Jasper
- US Army Medical Materiel Development Activity, Frederick, Maryland, USA
| | - Timothy P Endy
- SUNY Upstate Medical University, Institute for Global Health and Translational Sciences, Syracuse, New York, USA
| | - Richard G Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Stephen J Thomas
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| |
Collapse
|
16
|
Dayan GH, Langevin E, Forrat R, Zambrano B, Noriega F, Frago C, Bouckenooghe A, Machabert T, Savarino S, DiazGranados CA. Efficacy after 1 and 2 doses of CYD-TDV in dengue endemic areas by dengue serostatus. Vaccine 2020; 38:6472-6477. [DOI: 10.1016/j.vaccine.2020.07.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/13/2020] [Accepted: 07/26/2020] [Indexed: 01/21/2023]
|
17
|
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
|
18
|
Laher F, Bekker LG, Garrett N, Lazarus EM, Gray GE. Review of preventative HIV vaccine clinical trials in South Africa. Arch Virol 2020; 165:2439-2452. [PMID: 32797338 PMCID: PMC7426202 DOI: 10.1007/s00705-020-04777-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
New HIV infections continue relentlessly in southern Africa, demonstrating
the need for a vaccine to prevent HIV subtype C. In South Africa, the country with the
highest number of new infections annually, HIV vaccine research has been ongoing since
2003 with collaborative public-private-philanthropic partnerships. So far, 21 clinical
trials have been conducted in South Africa, investigating seven viral vectors, three DNA
plasmids, four envelope proteins, five adjuvants and three monoclonal antibodies. Active
vaccine candidates have spanned subtypes A, B, C, E and multi-subtype mosaic sequences.
All were well tolerated. Four concepts were investigated for efficacy: rAd5-gag/pol/nef
showed increased HIV acquisition in males, subtype C ALVAC/gp120/MF59 showed no
preventative efficacy, and the trials for the VRC01 monoclonal antibody and
Ad26.Mos4.HIV/subtype C gp140/ aluminum phosphate are ongoing. Future trials are planned
with DNA/viral vector plus protein combinations in concert with pre-exposure
prophylaxis, and sequential immunization studies with transmitted/founder HIV envelope
to induce broadly neutralizing antibodies. Finally, passive immunization trials are
underway to build on the experience with VRC01, including single and combination
antibody trials with an antibody derived from a subtype-C-infected South African donor.
Future consideration should be given to the evaluation of novel strategies, for example,
inactivated-whole-virus vaccines.
Collapse
Affiliation(s)
- Fatima Laher
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Linda-Gail Bekker
- The Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa.,Department of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
| | - Erica M Lazarus
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Glenda E Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
19
|
Liu R, Wang Y, Li J, Han H, Xia Z, Liu F, Wu K, Yang L, Liu X, Zhu C. Decreased T cell populations contribute to the increased severity of COVID-19. Clin Chim Acta 2020; 508:110-114. [PMID: 32405080 PMCID: PMC7219428 DOI: 10.1016/j.cca.2020.05.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND We observe changes of the main lymphocyte subsets (CD16+CD56、CD19、CD3、CD4、and CD8) in COVID-19-infected patients and explore whether the changes are associated with disease severity. METHODS One-hundred and fifty-four cases of COVID-19-infected patients were selected and divided into 3 groups (moderate group, severe group and critical group). The flow cytometry assay was performed to examine the numbers of lymphocyte subsets. RESULTS CD3+, CD4+ and CD8 + T lymphocyte subsets were decreased in COVID-19-infected patients. Compared with the moderate group and the sever group, CD3+, CD4+ and CD8+ T cells in the critical group decreased greatly (P < 0.001, P = 0.005 or P = 0.001). CONCLUSIONS Reduced CD3+, CD4+, CD8+ T lymphocyte counts may reflect the severity of the COVID-19. Monitoring T cell changes has important implications for the diagnosis and treatment of severe patients who may become critically ill.
Collapse
Affiliation(s)
- Rui Liu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Ying Wang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Sino-French Cooperative Central Lab, Shanghai Pudong Gongli Hospital, Secondary Military Medical University, Shanghai 200135, PR China
| | - Jie Li
- Postgraduate training base in Shanghai Gongli Hospital, Ningxia medical university, Pudong New Area, Shanghai200135, PR China
| | - Huan Han
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Zunen Xia
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Fang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, PR China; Wuhan Institute of Biotechnology, Wuhan, Hubei 430075, PR China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Lan Yang
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai200135, PR China
| | - Xinghui Liu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai200135, PR China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| |
Collapse
|