701
|
Ulanday GEL, Okamoto K, Morita K. Development and utility of an in vitro, fluorescence-based assay for the discovery of novel compounds against dengue 2 viral protease. Trop Med Health 2016; 44:22. [PMID: 27551237 PMCID: PMC4979183 DOI: 10.1186/s41182-016-0025-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/18/2016] [Indexed: 11/27/2022] Open
Abstract
Background Dengue disease is one of the most significant vector-borne illnesses in the world. The emergence and re-emergence of dengue infections in many parts of the world affect millions annually and continue to burden public health systems especially in low-income populations. Advances in dengue vaccine development showed promising results; however, protection seems to be suboptimal. There is no licensed chemotherapeutic agent against dengue to date. An ideal scenario of combinatorial vaccination of high-risk individuals and chemotherapy of the diseased during outbreaks may compensate for the meager protection offered by the vaccine. The dengue virus protease is important to viral replication and, as such, has been identified as a potential target for antivirals. It is, therefore, our objective to establish and optimize an appropriate screening method for use during the early stages of drug development for dengue. Methods In this study, we developed and optimized a biochemical assay system for use in screening compound libraries against dengue virus protease. We tested the selected protease inhibitors with a cell-based assay to determine inhibition of viral replication. Results We have presented direct plots of substrate kinetics data showing an apparent inhibition of the protease at excessive substrate concentrations. The most common sources of interference that may have affected the said observation were elucidated. Finally, a screen was done on an existing compound library using the developed method. The compounds selected in this study showed inhibitory activity against both the recombinant dengue protease and cell-based infectivity assays. Conclusions Our study shows the practicality of a customized biochemical assay to find possible inhibitors of dengue viral protease during the initial stages of drug discovery. Electronic supplementary material The online version of this article (doi:10.1186/s41182-016-0025-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gianne Eduard L Ulanday
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ; Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kenta Okamoto
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ; Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan ; Leading Graduate School Program, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
702
|
Abstract
Dengue is widespread throughout the tropics and local spatial variation in dengue virus transmission is strongly influenced by rainfall, temperature, urbanization and distribution of the principal mosquito vector Aedes aegypti. Currently, endemic dengue virus transmission is reported in the Eastern Mediterranean, American, South-East Asian, Western Pacific and African regions, whereas sporadic local transmission has been reported in Europe and the United States as the result of virus introduction to areas where Ae. aegypti and Aedes albopictus, a secondary vector, occur. The global burden of the disease is not well known, but its epidemiological patterns are alarming for both human health and the global economy. Dengue has been identified as a disease of the future owing to trends toward increased urbanization, scarce water supplies and, possibly, environmental change. According to the WHO, dengue control is technically feasible with coordinated international technical and financial support for national programmes. This Primer provides a general overview on dengue, covering epidemiology, control, disease mechanisms, diagnosis, treatment and research priorities.
Collapse
Affiliation(s)
- Maria G Guzman
- Institute of Tropical Medicine 'Pedro Kouri', PAHO/WHO Collaborating Center for the Study of Dengue and its Vector, Autopista Novia del Mediodia, Km 6 1/2, Havana 11400, Cuba
| | - Duane J Gubler
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Alienys Izquierdo
- Institute of Tropical Medicine 'Pedro Kouri', PAHO/WHO Collaborating Center for the Study of Dengue and its Vector, Autopista Novia del Mediodia, Km 6 1/2, Havana 11400, Cuba
| | - Eric Martinez
- Institute of Tropical Medicine 'Pedro Kouri', PAHO/WHO Collaborating Center for the Study of Dengue and its Vector, Autopista Novia del Mediodia, Km 6 1/2, Havana 11400, Cuba
| | - Scott B Halstead
- Department of Preventive Medicine and Biometrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
703
|
|
704
|
Andrade CC, Young KI, Johnson WL, Villa ME, Buraczyk CA, Messer WB, Hanley KA. Rise and fall of vector infectivity during sequential strain displacements by mosquito-borne dengue virus. J Evol Biol 2016; 29:2205-2218. [PMID: 27500505 DOI: 10.1111/jeb.12939] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/10/2016] [Accepted: 06/15/2016] [Indexed: 12/31/2022]
Abstract
Each of the four serotypes of mosquito-borne dengue virus (DENV-1-4) comprises multiple, genetically distinct strains. Competitive displacement between strains within a serotype is a common feature of DENV epidemiology and can trigger outbreaks of dengue disease. We investigated the mechanisms underlying two sequential displacements by DENV-3 strains in Sri Lanka that each coincided with abrupt increases in dengue haemorrhagic fever (DHF) incidence. First, the post-DHF strain displaced the pre-DHF strain in the 1980s. We have previously shown that post-DHF is more infectious than pre-DHF for the major DENV vector, Aedes aegypti. Then, the ultra-DHF strain evolved in situ from post-DHF and displaced its ancestor in the 2000s. We predicted that ultra-DHF would be more infectious for Ae. aegypti than post-DHF but found that ultra-DHF infected a significantly lower percentage of mosquitoes than post-DHF. We therefore hypothesized that ultra-DHF had effected displacement by disseminating in Ae. aegypti more rapidly than post-DHF, but this was not borne out by a time course of mosquito infection. To elucidate the mechanisms that shape these virus-vector interactions, we tested the impact of RNA interference (RNAi), the principal mosquito defence against DENV, on replication of each of the three DENV strains. Replication of all strains was similar in mosquito cells with dysfunctional RNAi, but in cells with functional RNAi, replication of pre-DHF was significantly suppressed relative to the other two strains. Thus, differences in susceptibility to RNAi may account for the differences in mosquito infectivity between pre-DHF and post-DHF, but other mechanisms underlie the difference between post-DHF and ultra-DHF.
Collapse
Affiliation(s)
- C C Andrade
- Department of Biology, New Mexico State University, Las Cruces, NM, USA.
| | - K I Young
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - W L Johnson
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - M E Villa
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - C A Buraczyk
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - W B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR, USA.,Division of Infectious Diseases, Department of Medicine, Oregon Health and Sciences University, Portland, OR, USA
| | - K A Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
705
|
Halstead SB. Critique of World Health Organization Recommendation of a Dengue Vaccine. J Infect Dis 2016; 214:1793-1795. [PMID: 27496975 DOI: 10.1093/infdis/jiw340] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/07/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Scott B Halstead
- Department of Preventive Medicine and Biometrics, Uniformed Services University of the Health Sciences, Rockville, Maryland
| |
Collapse
|
706
|
Wilder-Smith A, Vannice KS, Hombach J, Farrar J, Nolan T. Population Perspectives and World Health Organization Recommendations for CYD-TDV Dengue Vaccine. J Infect Dis 2016; 214:1796-1799. [DOI: 10.1093/infdis/jiw341] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 11/13/2022] Open
|
707
|
Jit M. The economic burden of dengue: no longer invisible or unavoidable. THE LANCET. INFECTIOUS DISEASES 2016; 16:873-4. [DOI: 10.1016/s1473-3099(16)30001-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 10/21/2022]
|
708
|
Hadinegoro SRS, Arredondo-García JL, Guy B, Bouckenooghe A, Noriega F, Jackson N. Answer to the review from Halstead and Russell “Protective and immunological behavior of chimeric yellow fever dengue vaccine” (DOI 10.1016/j.vaccine.2016.02.004). Vaccine 2016; 34:4273-4. [DOI: 10.1016/j.vaccine.2016.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/27/2016] [Accepted: 06/01/2016] [Indexed: 11/26/2022]
|
709
|
Ewer KJ, Lambe T, Rollier CS, Spencer AJ, Hill AVS, Dorrell L. Viral vectors as vaccine platforms: from immunogenicity to impact. Curr Opin Immunol 2016; 41:47-54. [DOI: 10.1016/j.coi.2016.05.014] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 11/29/2022]
|
710
|
Sung C, Wei Y, Watanabe S, Lee HS, Khoo YM, Fan L, Rathore APS, Chan KWK, Choy MM, Kamaraj US, Sessions OM, Aw P, de Sessions PF, Lee B, Connolly JE, Hibberd ML, Vijaykrishna D, Wijaya L, Ooi EE, Low JGH, Vasudevan SG. Extended Evaluation of Virological, Immunological and Pharmacokinetic Endpoints of CELADEN: A Randomized, Placebo-Controlled Trial of Celgosivir in Dengue Fever Patients. PLoS Negl Trop Dis 2016; 10:e0004851. [PMID: 27509020 PMCID: PMC4980036 DOI: 10.1371/journal.pntd.0004851] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED CELADEN was a randomized placebo-controlled trial of 50 patients with confirmed dengue fever to evaluate the efficacy and safety of celgosivir (A study registered at ClinicalTrials.gov, number NCT01619969). Celgosivir was given as a 400 mg loading dose and 200 mg bid (twice a day) over 5 days. Replication competent virus was measured by plaque assay and compared to reverse transcription quantitative PCR (qPCR) of viral RNA. Pharmacokinetics (PK) correlations with viremia, immunological profiling, next generation sequence (NGS) analysis and hematological data were evaluated as exploratory endpoints here to identify possible signals of pharmacological activity. Viremia by plaque assay strongly correlated with qPCR during the first four days. Immunological profiling demonstrated a qualitative shift in T helper cell profile during the course of infection. NGS analysis did not reveal any prominent signature that could be associated with drug treatment; however the phylogenetic spread of patients' isolates underlines the importance of strain variability that may potentially confound interpretation of dengue drug trials conducted during different outbreaks and in different countries. Celgosivir rapidly converted to castanospermine (Cast) with mean peak and trough concentrations of 5727 ng/mL (30.2 μM) and 430 ng/mL (2.3 μM), respectively and cleared with a half-life of 2.5 (± 0.6) hr. Mean viral log reduction between day 2 and 4 (VLR2-4) was significantly greater in secondary dengue than primary dengue (p = 0.002). VLR2-4 did not correlate with drug AUC but showed a trend of greater response with increasing Cmin. PK modeling identified dosing regimens predicted to achieve 2.4 to 4.5 times higher Cmin. than in the CELADEN trial for only 13% to 33% increase in overall dose. A small, non-statistical trend towards better outcome on platelet nadir and difference between maximum and minimum hematocrit was observed in celgosivir-treated patients with secondary dengue infection. Optimization of the dosing regimen and patient stratification may enhance the ability of a clinical trial to demonstrate celgosivir activity in treating dengue fever based on hematological endpoints. A new clinical trial with a revised dosing regimen is slated to start in 2016 (NCT02569827). Furthermore celgosivir's potential value for treatment of other flaviruses such as Zika virus should be investigated urgently. TRIAL REGISTRATION ClinicalTrials.gov NCT01619969.
Collapse
Affiliation(s)
- Cynthia Sung
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Vigilance and Compliance Branch, Health Sciences Authority, Singapore
| | - Yuan Wei
- Singapore Clinical Research Institute, Singapore
| | - Satoru Watanabe
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - How Sung Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yok Moi Khoo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lu Fan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Kitti Wing-Ki Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Milly M. Choy
- Genome Institute of Singapore, A*STAR, Biopolis, Singapore
| | - Uma S. Kamaraj
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Pauline Aw
- Genome Institute of Singapore, A*STAR, Biopolis, Singapore
| | | | - Bernett Lee
- Institute of Molecular & Cellular Biology, A*STAR, Biopolis, Singapore
| | - John E. Connolly
- Institute of Molecular & Cellular Biology, A*STAR, Biopolis, Singapore
| | | | | | - Limin Wijaya
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | | |
Collapse
|
711
|
Aguiar M, Stollenwerk N, Halstead SB. The risks behind Dengvaxia recommendation. THE LANCET. INFECTIOUS DISEASES 2016; 16:882-3. [DOI: 10.1016/s1473-3099(16)30168-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/06/2016] [Indexed: 11/15/2022]
|
712
|
Nealon J, Taurel AF, Capeding MR, Tran NH, Hadinegoro SR, Chotpitayasunondh T, Chong CK, Wartel TA, Beucher S, Frago C, Moureau A, Simmerman M, Laot T, L’Azou M, Bouckenooghe A. Symptomatic Dengue Disease in Five Southeast Asian Countries: Epidemiological Evidence from a Dengue Vaccine Trial. PLoS Negl Trop Dis 2016; 10:e0004918. [PMID: 27532617 PMCID: PMC4988713 DOI: 10.1371/journal.pntd.0004918] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/22/2016] [Indexed: 12/03/2022] Open
Abstract
Dengue incidence has increased globally, but empirical burden estimates are scarce. Prospective methods are best-able to capture all severities of disease. CYD14 was an observer-blinded dengue vaccine study conducted in children 2-14 years of age in Indonesia, Malaysia, Thailand, the Philippines, and Vietnam. The control group received no vaccine and resembled a prospective, observational study. We calculated the rates of dengue according to different laboratory or clinical criteria to make inferences about dengue burden, and compared with rates reported in the passive surveillance systems to calculate expansion factors which describe under-reporting. Over 6,933 person-years of observation in the control group there were 319 virologically confirmed dengue cases, a crude attack rate of 4.6%/year. Of these, 92 cases (28.8%) were clinically diagnosed as dengue fever or dengue hemorrhagic fever by investigators and 227 were not, indicating that most symptomatic disease fails to satisfy existing case definitions. When examining different case definitions, there was an inverse relationship between clinical severity and observed incidence rates. CYD14's active surveillance system captured a greater proportion of symptomatic dengue than national passive surveillance systems, giving rise to expansion factors ranging from 0.5 to 31.7. This analysis showed substantial, unpredictable and variable under-reporting of symptomatic dengue, even within a controlled clinical trial environment, and emphasizes that burden estimates are highly sensitive to case definitions. These data will assist in generating disease burden estimates and have important policy implications when considering the introduction and health economics of dengue prevention and control interventions.
Collapse
Affiliation(s)
| | | | | | - Ngoc Huu Tran
- Pasteur Institute Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Sri Rezeki Hadinegoro
- University of Indonesia Medical School, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | | | - Chee Kheong Chong
- Disease Control Division, Malaysian Ministry of Health, Putrajaya, Malaysia
| | | | | | | | | | | | - Thelma Laot
- Sanofi Pasteur Asia & JPAC Region, Singapore
| | - Maïna L’Azou
- Sanofi Pasteur, Global Epidemiology, Lyon, France
| | | |
Collapse
|
713
|
Lim SK, Lee YS, Namkung S, Lim JK, Yoon IK. Prospects for dengue vaccines for travelers. Clin Exp Vaccine Res 2016; 5:89-100. [PMID: 27489798 PMCID: PMC4969283 DOI: 10.7774/cevr.2016.5.2.89] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/19/2022] Open
Abstract
Travel-acquired dengue cases have been increasing as the overall global dengue burden has expanded. In Korea, imported dengue cases have been reported since 2000 when it first became a notifiable disease. During the first four months of 2016, three times more dengue cases were reported in Korea than during the same period the previous year. A safe and efficacious vaccine for travelers would be beneficial to prevent dengue disease in individual travelers and potentially decrease the risk of virus spread to non-endemic areas. Here, we summarize the characteristics of dengue vaccines for travelers and review dengue vaccines currently licensed or in clinical development.
Collapse
Affiliation(s)
- Sl-Ki Lim
- International Vaccine Institute, Seoul, Korea
| | | | - Suk Namkung
- International Vaccine Institute, Seoul, Korea
| | | | - In-Kyu Yoon
- International Vaccine Institute, Seoul, Korea
| |
Collapse
|
714
|
Chiang CY, Pan CH, Chen MY, Hsieh CH, Tsai JP, Liu HH, Liu SJ, Chong P, Leng CH, Chen HW. Immunogenicity of a novel tetravalent vaccine formulation with four recombinant lipidated dengue envelope protein domain IIIs in mice. Sci Rep 2016; 6:30648. [PMID: 27470096 PMCID: PMC4965760 DOI: 10.1038/srep30648] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/08/2016] [Indexed: 11/09/2022] Open
Abstract
We developed a novel platform to express high levels of recombinant lipoproteins with intrinsic adjuvant properties. Based on this technology, our group developed recombinant lipidated dengue envelope protein domain IIIs as vaccine candidates against dengue virus. This work aims to evaluate the immune responses in mice to the tetravalent formulation. We demonstrate that 4 serotypes of recombinant lipidated dengue envelope protein domain III induced both humoral and cellular immunity against all 4 serotypes of dengue virus on the mixture that formed the tetravalent formulation. Importantly, the immune responses induced by the tetravalent formulation in the absence of the exogenous adjuvant were functional in clearing the 4 serotypes of dengue virus in vivo. We affirm that the tetravalent formulation of recombinant lipidated dengue envelope protein domain III is a potential vaccine candidate against dengue virus and suggest further detailed studies of this formulation in nonhuman primates.
Collapse
Affiliation(s)
- Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
| | - Chien-Hsiung Pan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, Republic of China
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
| | - Chun-Hsiang Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
| | - Jy-Ping Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
| | - Hsueh-Hung Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, Republic of China
| | - Pele Chong
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, Republic of China
| | - Chih-Hsiang Leng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, Republic of China
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, Republic of China
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
715
|
Cytokine Profile of Children Hospitalized with Virologically-Confirmed Dengue during Two Phase III Vaccine Efficacy Trials. PLoS Negl Trop Dis 2016; 10:e0004830. [PMID: 27459266 PMCID: PMC4961416 DOI: 10.1371/journal.pntd.0004830] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/17/2016] [Indexed: 01/29/2023] Open
Abstract
Background Two large-scale efficacy studies with the recombinant yellow fever-17D–dengue virus, live-attenuated, tetravalent dengue vaccine (CYD-TDV) candidate undertaken in Asia (NCT01373281) and Latin America (NCT01374516) demonstrated significant protection against dengue disease during two years’ active surveillance (active phase). Long-term follow up of participants for breakthrough disease leading to hospitalization is currently ongoing (hospital phase). Methodology/Principal findings We assessed the cytokine profile in acute sera from selected participants hospitalized (including during the active phase) up to the beginning of the second year of long-term follow up for both studies. The serum concentrations of 38 cytokines were measured in duplicate using the Milliplex Human Cytokine MAGNETIC BEAD Premixed 38 Plex commercial kit (Millipore, Billerica, MA, USA). Partial least squares discriminant analyses did not reveal any difference in the overall cytokine profile of CYD-TDV and placebo recipients hospitalized for breakthrough dengue regardless of stratification used. In addition, there was no difference in the cytokine profile for breakthrough dengue among those aged <9 years versus those aged ≥ 9 years. Conclusions/Significance These exploratory findings show that CYD-TDV does not induce a particular immune profile versus placebo, corroborating the clinical profile observed. A live-attenuated, tetravalent dengue vaccine (CYD-TDV) has been shown to provide protection against dengue disease in two large-scale, placebo-controlled, phase III efficacy studies. Continued surveillance of study participants was subsequently undertaken to better define longer term vaccine efficacy and safety. A yet unexplained higher incidence of hospitalization for dengue disease was observed among children aged <9 years in year 3 of follow up. While the clinical outcome of the hospitalized cases was similar between CYD-TDV and placebo recipients, it was important to further investigate whether the immune profile induced by breakthrough infection differed between the two study groups. We compared the profile of 38 cytokines, chemokines and growth factors in acute phase sera collected from participants with breakthrough disease in the two groups. No difference in overall profile was observed between CYD-TDV and placebo recipients. Similarly, no difference in the cytokine profile for breakthrough dengue was observed between those aged <9 years and those aged ≥ 9 years. Based on these analyzed factors, our study shows that CYD-TDV does not induce an overall altered immunological profile with breakthrough disease compared with placebo, in agreement with the similar clinical pictures and viremia observed in the two groups.
Collapse
|
716
|
Assessing dengue vaccination impact: Model challenges and future directions. Vaccine 2016; 34:4461-4465. [PMID: 27461457 DOI: 10.1016/j.vaccine.2016.06.082] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/23/2016] [Accepted: 06/29/2016] [Indexed: 11/23/2022]
Abstract
In response to the sharp rise in the global burden caused by dengue virus (DENV) over the last few decades, the WHO has set out three specific key objectives in its disease control strategy: (i) to estimate the true burden of dengue by 2015; (ii) a reduction in dengue mortality by at least 50% by 2020 (used as a baseline); and (iii) a reduction in dengue morbidity by at least 25% by 2020. Although various elements will all play crucial parts in achieving this goal, from diagnosis and case management to integrated surveillance and outbreak response, sustainable vector control, vaccine implementation and finally operational and implementation research, it seems clear that new tools (e.g. a safe and effective vaccine and/or effective vector control) are key to success. The first dengue vaccine was licensed in December 2015, Dengvaxia® (CYD-TDV) developed by Sanofi Pasteur. The WHO has provided guidance on the use of CYD-TDV in endemic countries, for which there are a variety of considerations beyond the risk-benefit evaluation done by regulatory authorities, including public health impact and cost-effectiveness. Population-level vaccine impact and economic and financial aspects are two issues that can potentially be considered by means of mathematical modelling, especially for new products for which empirical data are still lacking. In December 2014 a meeting was convened by the WHO in order to revisit the current status of dengue transmission models and their utility for public health decision-making. Here, we report on the main points of discussion and the conclusions of this meeting, as well as next steps for maximising the use of mathematical models for vaccine decision-making.
Collapse
|
717
|
Everts M, Cihlar T, Bostwick JR, Whitley RJ. Accelerating Drug Development: Antiviral Therapies for Emerging Viruses as a Model. Annu Rev Pharmacol Toxicol 2016; 57:155-169. [PMID: 27483339 DOI: 10.1146/annurev-pharmtox-010716-104533] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Drug discovery and development is a lengthy and expensive process. Although no one, simple, single solution can significantly accelerate this process, steps can be taken to avoid unnecessary delays. Using the development of antiviral therapies as a model, we describe options for acceleration that cover target selection, assay development and high-throughput screening, hit confirmation, lead identification and development, animal model evaluations, toxicity studies, regulatory issues, and the general drug discovery and development infrastructure. Together, these steps could result in accelerated timelines for bringing antiviral therapies to market so they can treat emerging infections and reduce human suffering.
Collapse
Affiliation(s)
- Maaike Everts
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama, Birmingham, Alabama 35233; ,
| | - Tomas Cihlar
- Department of Biology, Gilead Sciences, Inc., Foster City, California 94404;
| | - J Robert Bostwick
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205;
| | - Richard J Whitley
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama, Birmingham, Alabama 35233; ,
| |
Collapse
|
718
|
Liu Y, Liu J, Cheng G. Vaccines and immunization strategies for dengue prevention. Emerg Microbes Infect 2016; 5:e77. [PMID: 27436365 PMCID: PMC5141265 DOI: 10.1038/emi.2016.74] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/20/2016] [Accepted: 05/11/2016] [Indexed: 01/01/2023]
Abstract
Dengue is currently the most significant arboviral disease afflicting tropical and sub-tropical countries worldwide. Dengue vaccines, such as the multivalent attenuated, chimeric, DNA and inactivated vaccines, have been developed to prevent dengue infection in humans, and they function predominantly by stimulating immune responses against the dengue virus (DENV) envelope (E) and nonstructural-1 proteins (NS1). Of these vaccines, a live attenuated chimeric tetravalent DENV vaccine developed by Sanofi Pasteur has been licensed in several countries. However, this vaccine renders only partial protection against the DENV2 infection and is associated with an unexplained increased incidence of hospitalization for severe dengue disease among children younger than nine years old. In addition to the virus-based vaccines, several mosquito-based dengue immunization strategies have been developed to interrupt the vector competence and effectively reduce the number of infected mosquito vectors, thus controlling the transmission of DENV in nature. Here we summarize the recent progress in the development of dengue vaccines and novel immunization strategies and propose some prospective vaccine strategies for disease prevention in the future.
Collapse
Affiliation(s)
- Yang Liu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.,School of Life Science, Tsinghua University, Beijing 100084, China
| | - Jianying Liu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
719
|
Khetarpal N, Khanna I. Dengue Fever: Causes, Complications, and Vaccine Strategies. J Immunol Res 2016; 2016:6803098. [PMID: 27525287 PMCID: PMC4971387 DOI: 10.1155/2016/6803098] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/18/2016] [Accepted: 06/01/2016] [Indexed: 01/19/2023] Open
Abstract
Dengue is a highly endemic infectious disease of the tropical countries and is rapidly becoming a global burden. It is caused by any of the 4 serotypes of dengue virus and is transmitted within humans through female Aedes mosquitoes. Dengue disease varies from mild fever to severe conditions of dengue hemorrhagic fever and shock syndrome. Globalization, increased air travel, and unplanned urbanization have led to increase in the rate of infection and helped dengue to expand its geographic and demographic distribution. Dengue vaccine development has been a challenging task due to the existence of four antigenically distinct dengue virus serotypes, each capable of eliciting cross-reactive and disease-enhancing antibody response against the remaining three serotypes. Recently, Sanofi Pasteur's chimeric live-attenuated dengue vaccine candidate has been approved in Mexico, Brazil, and Philippines for usage in adults between 9 and 45 years of age. The impact of its limited application to the public health system needs to be evaluated. Simultaneously, the restricted application of this vaccine candidate warrants continued efforts in developing a dengue vaccine candidate which is additionally efficacious for infants and naïve individuals. In this context, alternative strategies of developing a designed vaccine candidate which does not allow production of enhancing antibodies should be explored, as it may expand the umbrella of efficacy to include infants and naïve individuals.
Collapse
Affiliation(s)
- Niyati Khetarpal
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Department of Biochemistry, University of Delhi, Institute of Home Economics, Hauz Khas, New Delhi 110016, India
| | - Ira Khanna
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
720
|
Affiliation(s)
| | - Maira Aguiar
- Center for Mathematics, Fundamental Applications and Operations Research, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
721
|
Olivera-Botello G, Coudeville L, Fanouillere K, Guy B, Chambonneau L, Noriega F, Jackson N. Tetravalent Dengue Vaccine Reduces Symptomatic and Asymptomatic Dengue Virus Infections in Healthy Children and Adolescents Aged 2-16 Years in Asia and Latin America. J Infect Dis 2016; 214:994-1000. [PMID: 27418050 PMCID: PMC5021228 DOI: 10.1093/infdis/jiw297] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/11/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Asymptomatic dengue virus-infected individuals are thought to play a major role in dengue virus transmission. The efficacy of the recently approved quadrivalent CYD-TDV dengue vaccine against asymptomatic dengue virus infection has not been previously assessed. METHODS We pooled data for 3 736 individuals who received either CYD-TDV or placebo at 0, 6, and 12 months in the immunogenicity subsets of 2 phase 3 trials (clinical trials registration NCT01373281 and NCT01374516). We defined a seroconversion algorithm (ie, a ≥4-fold increase in the neutralizing antibody titer and a titer of ≥40 from month 13 to month 25) as a surrogate marker of asymptomatic infection in the vaccine and placebo groups. RESULTS The algorithm detected seroconversion in 94% of individuals with a diagnosis of virologically confirmed dengue between months 13 and 25, validating its discriminatory power. Among those without virologically confirmed dengue (n = 3 669), 219 of 2 485 in the vaccine group and 157 of 1 184 in the placebo group seroconverted between months 13 and 25, giving a vaccine efficacy of 33.5% (95% confidence interval [CI], 17.9%-46.1%) against asymptomatic infection. Vaccine efficacy was marginally higher in subjects aged 9-16 years (38.6%; 95% CI, 22.1%-51.5%). The annual incidence of asymptomatic dengue virus infection in this age group was 14.8%, which was 4.4 times higher than the incidence for symptomatic dengue (3.4%). CONCLUSIONS The observed vaccine efficacy against asymptomatic dengue virus infections is expected to translate into reduced dengue virus transmission if sufficient individuals are vaccinated in dengue-endemic areas.
Collapse
|
722
|
Gailhardou S, Skipetrova A, Dayan GH, Jezorwski J, Saville M, Van der Vliet D, Wartel TA. Safety Overview of a Recombinant Live-Attenuated Tetravalent Dengue Vaccine: Pooled Analysis of Data from 18 Clinical Trials. PLoS Negl Trop Dis 2016; 10:e0004821. [PMID: 27414655 PMCID: PMC4945086 DOI: 10.1371/journal.pntd.0004821] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/12/2016] [Indexed: 11/19/2022] Open
Abstract
A recombinant live attenuated tetravalent dengue vaccine (CYD-TDV) has been shown to be efficacious in preventing virologically-confirmed dengue disease, severe dengue disease and dengue hospitalization in children aged 2-16 years in Asia and Latin America. We analyzed pooled safety data from 18 phase I, II and III clinical trials in which the dengue vaccine was administered to participants aged 2-60 years, including long-term safety follow-up in three efficacy trials. The participants were analyzed according to their age at enrollment. The percentage of participants aged 2-60 years reporting ≥1 solicited injection-site or systemic reactions was slightly higher in the CYD-TDV group than in the placebo group. The most common solicited injection-site reactions were pain. Headache and malaise were the most common solicited systemic reactions. In both groups 0.3% of participants discontinued for safety reasons. The most common unsolicited adverse events were injection-site reactions, gastrointestinal disorders, and infections. Reactogenicity did not increase with successive doses of CYD-TDV. The frequency and nature of SAEs occurring within 28 days of any dose were similar in the CYD-TDV and placebo groups and were common medical conditions that could be expected as a function of age. Baseline dengue virus serostatus did not appear to influence the safety profile. No vaccine-related anaphylactic reactions, neurotropic events or viscerotropic events were reported. In year 3 after dose 1, an imbalance for dengue hospitalization, including for severe dengue, observed in participants aged <9 years in the CYD-TDV group compared with the placebo group was not observed for participants aged ≥9 years. In Year 4, this imbalance in participants aged <9 years was less marked, giving an overall lower risk of dengue hospitalization or severe dengue from dose 1 to Year 4 in the CYD-TDV group. These results have contributed to the definition of the target population for vaccination (≥9 years old) for which CYD-TDV has a satisfactory safety profile. Long-term safety will continue to be monitored in the ongoing follow-up of efficacy trials. Safety and effectiveness in real-life settings will be assessed through post-licensure studies.
Collapse
Affiliation(s)
| | | | - Gustavo H. Dayan
- Sanofi Pasteur, Swiftwater, Pennsylvania, United States of America
| | - John Jezorwski
- Sanofi Pasteur, Swiftwater, Pennsylvania, United States of America
| | | | | | | |
Collapse
|
723
|
Gottschamel J, Lössl A, Ruf S, Wang Y, Skaugen M, Bock R, Clarke JL. Production of dengue virus envelope protein domain III-based antigens in tobacco chloroplasts using inducible and constitutive expression systems. PLANT MOLECULAR BIOLOGY 2016; 91:497-512. [PMID: 27116001 DOI: 10.1007/s11103-016-0484-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 04/17/2016] [Indexed: 06/05/2023]
Abstract
Dengue fever is a disease in many parts of the tropics and subtropics and about half the world's population is at risk of infection according to the World Health Organization. Dengue is caused by any of the four related dengue virus serotypes DEN-1, -2, -3 and -4, which are transmitted to people by Aedes aegypti mosquitoes. Currently there is only one vaccine (Dengvaxia(®)) available (limited to a few countries) on the market since 2015 after half a century's intensive efforts. Affordable and accessible vaccines against dengue are hence still urgently needed. The dengue envelop protein domain III (EDIII), which is capable of eliciting serotype-specific neutralizing antibodies, has become the focus for subunit vaccine development. To contribute to the development of an accessible and affordable dengue vaccine, in the current study we have used plant-based vaccine production systems to generate a dengue subunit vaccine candidate in tobacco. Chloroplast genome engineering was applied to express serotype-specific recombinant EDIII proteins in tobacco chloroplasts using both constitutive and ethanol-inducible expression systems. Expression of a tetravalent antigen fusion construct combining EDIII polypeptides from all four serotypes was also attempted. Transplastomic EDIII-expressing tobacco lines were obtained and homoplasmy was verified by Southern blot analysis. Northern blot analyses showed expression of EDIII antigen-encoding genes. EDIII protein accumulation levels varied for the different recombinant EDIII proteins and the different expression systems, and reached between 0.8 and 1.6 % of total cellular protein. Our study demonstrates the suitability of the chloroplast compartment as a production site for an EDIII-based vaccine candidate against dengue fever and presents a Gateway(®) plastid transformation vector for inducible transgene expression.
Collapse
Affiliation(s)
- Johanna Gottschamel
- NIBIO-Norwegian Institute of Bioeconomy Research, P.O. Box 115, 1431, Ås, Norway
- BOKU-University of Natural Resources and Life Sciences, Gregor-Mendel-Straße 33, 1180, Vienna, Austria
| | - Andreas Lössl
- BOKU-University of Natural Resources and Life Sciences, Gregor-Mendel-Straße 33, 1180, Vienna, Austria
| | - Stephanie Ruf
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany
| | - Yanliang Wang
- NIBIO-Norwegian Institute of Bioeconomy Research, P.O. Box 115, 1431, Ås, Norway
| | | | - Ralph Bock
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany.
| | - Jihong Liu Clarke
- NIBIO-Norwegian Institute of Bioeconomy Research, P.O. Box 115, 1431, Ås, Norway.
| |
Collapse
|
724
|
Chemical diversity and antiviral potential in the pantropical Diospyros genus. Fitoterapia 2016; 112:9-15. [DOI: 10.1016/j.fitote.2016.04.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 01/02/2023]
|
725
|
Unique safety issues associated with virus-vectored vaccines: Potential for and theoretical consequences of recombination with wild type virus strains. Vaccine 2016; 34:6610-6616. [PMID: 27346303 PMCID: PMC5204448 DOI: 10.1016/j.vaccine.2016.04.060] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 04/20/2016] [Indexed: 12/26/2022]
Abstract
In 2003 and 2013, the World Health Organization convened informal consultations on characterization and quality aspects of vaccines based on live virus vectors. In the resulting reports, one of several issues raised for future study was the potential for recombination of virus-vectored vaccines with wild type pathogenic virus strains. This paper presents an assessment of this issue formulated by the Brighton Collaboration. To provide an appropriate context for understanding the potential for recombination of virus-vectored vaccines, we review briefly the current status of virus-vectored vaccines, mechanisms of recombination between viruses, experience with recombination involving live attenuated vaccines in the field, and concerns raised previously in the literature regarding recombination of virus-vectored vaccines with wild type virus strains. We then present a discussion of the major variables that could influence recombination between a virus-vectored vaccine and circulating wild type virus and the consequences of such recombination, including intrinsic recombination properties of the parent virus used as a vector; sequence relatedness of vector and wild virus; virus host range, pathogenesis and transmission; replication competency of vector in target host; mechanism of vector attenuation; additional factors potentially affecting virulence; and circulation of multiple recombinant vectors in the same target population. Finally, we present some guiding principles for vector design and testing intended to anticipate and mitigate the potential for and consequences of recombination of virus-vectored vaccines with wild type pathogenic virus strains.
Collapse
|
726
|
Halstead SB, Aguiar M. Dengue vaccines: Are they safe for travelers? Travel Med Infect Dis 2016; 14:378-83. [PMID: 27343438 DOI: 10.1016/j.tmaid.2016.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 10/21/2022]
Abstract
The four dengue viruses (DENV) circulate among nearly one-half of the world's population in tropical and semitropical countries imposing a huge morbidity burden on travelers. Sanofipasteur has developed a tetravalent live-attenuated vaccine, Dengvaxia, recently approved by the World Health Organization and licensed in four dengue-endemic countries. An additional two dengue vaccines, developed by the National Institute of Allergy and Infectious Diseases (NIAID), USA and Takeda, are entering phase III testing. Dengvaxia is composed of four yellow fever 17D-DENV chimeras, the NIAID vaccine contains three mutagenized DENV and one DENV2/4 chimera while the Takeda vaccine contains an attenuated DENV 2 and three DENV 2-DENV chimeras. Which of these vaccines might be useful in protecting travelers against dengue infections and disease? Dengvaxia requires three doses administered over the course of one year but in addition has safety signals suggesting that susceptible individuals should not be vaccinated. The NIAID vaccine is promising as a travel vaccine as a single dose fully protected susceptible adults against live dengue 2 virus challenge. The protective efficacy and safety of the Takeda vaccine remain to be demonstrated.
Collapse
Affiliation(s)
- Scott B Halstead
- Department of Preventive Medicine and Biometrics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Maira Aguiar
- BioMathematics and Statistics Group, Center for Mathematics, Fundamental Applications and Operations Research, Lisbon University, USA.
| |
Collapse
|
727
|
Pitisuttithum P, Bouckenooghe A. The first licensed dengue vaccine: an important tool for integrated preventive strategies against dengue virus infection. Expert Rev Vaccines 2016; 15:795-8. [DOI: 10.1080/14760584.2016.1189331] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Punnee Pitisuttithum
- Faculty of Tropical Medicine, Vaccine Trial Centre, Mahidol University, Bangkok, Thailand
| | - Alain Bouckenooghe
- Clinical Sciences and Medical Affairs Asia, Sanofi Pasteur, Singapore, Singapore
| |
Collapse
|
728
|
Will dengue vaccination be cost-effective for Argentina? Reply to letter by Urueña et al. regarding "Cost-utility analysis of dengue vaccination in a country with heterogeneous risk of dengue transmission". Vaccine 2016; 34:3221. [PMID: 27288996 DOI: 10.1016/j.vaccine.2016.03.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/30/2016] [Indexed: 11/21/2022]
|
729
|
Servín-Blanco R, Zamora-Alvarado R, Gevorkian G, Manoutcharian K. Antigenic variability: Obstacles on the road to vaccines against traditionally difficult targets. Hum Vaccin Immunother 2016; 12:2640-2648. [PMID: 27295540 DOI: 10.1080/21645515.2016.1191718] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Despite the impressive impact of vaccines on public health, the success of vaccines targeting many important pathogens and cancers has to date been limited. The burden of infectious diseases today is mainly caused by antigenically variable pathogens (AVPs), which escape immune responses induced by prior infection or vaccination through changes in molecular structures recognized by antibodies or T cells. Extensive genetic and antigenic variability is the major obstacle for the development of new or improved vaccines against "difficult" targets. Alternative, qualitatively new approaches leading to the generation of disease- and patient-specific vaccine immunogens that incorporate complex permanently changing epitope landscapes of intended targets accompanied by appropriate immunomodulators are urgently needed. In this review, we highlight some of the most critical common issues related to the development of vaccines against many pathogens and cancers that escape protective immune responses owing to antigenic variation, and discuss recent efforts to overcome the obstacles by applying alternative approaches for the rational design of new types of immunogens.
Collapse
Affiliation(s)
- R Servín-Blanco
- a Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria , México DF , México
| | - R Zamora-Alvarado
- a Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria , México DF , México
| | - G Gevorkian
- a Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria , México DF , México
| | - K Manoutcharian
- a Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Cuidad Universitaria , México DF , México
| |
Collapse
|
730
|
Yam-Puc JC, Cedillo-Barrón L, Aguilar-Medina EM, Ramos-Payán R, Escobar-Gutiérrez A, Flores-Romo L. The Cellular Bases of Antibody Responses during Dengue Virus Infection. Front Immunol 2016; 7:218. [PMID: 27375618 PMCID: PMC4893500 DOI: 10.3389/fimmu.2016.00218] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/20/2016] [Indexed: 11/26/2022] Open
Abstract
Dengue virus (DENV) is one of the most significant human viral pathogens transmitted by mosquitoes and can cause from an asymptomatic disease to mild undifferentiated fever, classical dengue, and severe dengue. Neutralizing memory antibody (Ab) responses are one of the most important mechanisms that counteract reinfections and are therefore the main aim of vaccination. However, it has also been proposed that in dengue, some of these class-switched (IgG) memory Abs might worsen the disease. Although these memory Abs derive from B cells by T-cell-dependent processes, we know rather little about the (acute, chronic, or memory) B cell responses and the complex cellular mechanisms generating these Abs during DENV infections. This review aims to provide an updated and comprehensive perspective of the B cell responses during DENV infection, starting since the very early events such as the cutaneous DENV entrance and the arrival into draining lymph nodes, to the putative B cell activation, proliferation, and germinal centers (GCs) formation (the source of affinity-matured class-switched memory Abs), till the outcome of GC reactions such as the generation of plasmablasts, Ab-secreting plasma cells, and memory B cells. We discuss topics very poorly explored such as the possibility of B cell infection by DENV or even activation-induced B cell death. The current information about the nature of the Ab responses to DENV is also illustrated.
Collapse
Affiliation(s)
- Juan Carlos Yam-Puc
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN , Mexico City , Mexico
| | - Leticia Cedillo-Barrón
- Department of Molecular Biomedicine, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN , Mexico City , Mexico
| | - Elsa Maribel Aguilar-Medina
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa (UAS) , Culiacan, Sinaloa , Mexico
| | - Rosalío Ramos-Payán
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa (UAS) , Culiacan, Sinaloa , Mexico
| | - Alejandro Escobar-Gutiérrez
- Department for Immunological Investigations, Institute for Epidemiological Diagnosis and Reference, Health Secretariat , Mexico City , Mexico
| | - Leopoldo Flores-Romo
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN , Mexico City , Mexico
| |
Collapse
|
731
|
Lourenço J, Recker M. Dengue serotype immune-interactions and their consequences for vaccine impact predictions. Epidemics 2016; 16:40-8. [PMID: 27663790 PMCID: PMC5030310 DOI: 10.1016/j.epidem.2016.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 05/31/2016] [Accepted: 05/31/2016] [Indexed: 11/11/2022] Open
Abstract
The firstever dengue vaccine, Dengvaxia®, has recently been licensed for use in several countries. Mathematical models are valuable tools for assessing vaccination impact on dengue burden. Model assumptions regarding dengue serotype immune interactions are inconsistent. Our results demonstrate how model assumptions critically affect vaccine impact predictions.
Dengue is one of the most important and wide-spread viral infections affecting human populations. The last few decades have seen a dramatic increase in the global burden of dengue, with the virus now being endemic or near-endemic in over 100 countries world-wide. A recombinant tetravalent vaccine candidate (CYD-TDV) has recently completed Phase III clinical efficacy trials in South East Asia and Latin America and has been licensed for use in several countries. The trial results showed moderate-to-high efficacies in protection against clinical symptoms and hospitalisation but with so far unknown effects on transmission and infections per se. Model-based predictions about the vaccine's short- or long-term impact on the burden of dengue are therefore subject to a considerable degree of uncertainty. Furthermore, different immune interactions between dengue's serotypes have frequently been evoked by modelling studies to underlie dengue's oscillatory dynamics in disease incidence and serotype prevalence. Here we show how model assumptions regarding immune interactions in the form of antibody-dependent enhancement, temporary cross-immunity and the number of infections required to develop full immunity can significantly affect the predicted outcome of a dengue vaccination campaign. Our results thus re-emphasise the important gap in our current knowledge concerning the effects of previous exposure on subsequent dengue infections and further suggest that intervention impact studies should be critically evaluated by their underlying assumptions about serotype immune-interactions.
Collapse
Affiliation(s)
- José Lourenço
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| | - Mario Recker
- Centre for Mathematics and the Environment, University of Exeter, Penryn Campus, Penryn TR10 9EZ, UK.
| |
Collapse
|
732
|
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]
|
733
|
Villabona-Arenas CJ, Ocazionez Jimenez RE, Jimenez Silva CL. Dengue Vaccine: Considerations before Rollout in Colombia. PLoS Negl Trop Dis 2016; 10:e0004653. [PMID: 27280803 PMCID: PMC4900517 DOI: 10.1371/journal.pntd.0004653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Christian Julian Villabona-Arenas
- UMI233, Institut de Recherche pour le Développement (IRD), Université de Montpellier, Montpellier, France
- Laboratoire d'Informatique, de Robotique et de Microélectronique de Montpellier (LIRMM), Institut de Biologie Computationnelle (IBC), Université de Montpellier, Montpellier, France
| | - Raquel Elvira Ocazionez Jimenez
- Laboratorio de Arbovirus, Centro de Investigaciones en Enfermedades Tropicales (CINTROP), Universidad Industrial de Santander, Santander, Colombia
| | - Cinthy Lorena Jimenez Silva
- Laboratorio de Arbovirus, Centro de Investigaciones en Enfermedades Tropicales (CINTROP), Universidad Industrial de Santander, Santander, Colombia
| |
Collapse
|
734
|
Urueña A, Vizzotti C, San Juan JA. Response to Orellano et al. regarding cost-utility analysis of dengue vaccination in a country with heterogeneous risk of dengue transmission. Vaccine 2016; 34:3222. [DOI: 10.1016/j.vaccine.2016.03.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/30/2016] [Indexed: 11/29/2022]
|
735
|
Reiner RC, Achee N, Barrera R, Burkot TR, Chadee DD, Devine GJ, Endy T, Gubler D, Hombach J, Kleinschmidt I, Lenhart A, Lindsay SW, Longini I, Mondy M, Morrison AC, Perkins TA, Vazquez-Prokopec G, Reiter P, Ritchie SA, Smith DL, Strickman D, Scott TW. Quantifying the Epidemiological Impact of Vector Control on Dengue. PLoS Negl Trop Dis 2016; 10:e0004588. [PMID: 27227829 PMCID: PMC4881945 DOI: 10.1371/journal.pntd.0004588] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Robert C. Reiner
- Department of Epidemiology and Biostatistics, Indiana University Bloomington School of Public Health, Bloomington, Indiana, United States of America
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Nicole Achee
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Roberto Barrera
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico
| | - Thomas R. Burkot
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Dave D. Chadee
- Department of Life Sciences, Faculty of Science and Agriculture, The University of the West Indies, St. Augustine Campus, St. Augustine, Trinidad and Tobago
| | - Gregor J. Devine
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Timothy Endy
- Department of Medicine, Upstate Medical University of New York, Syracuse, New York, United States of America
| | - Duane Gubler
- Signature Research Program in Emerging Infectious Disease, Duke-NUS Medical School, Singapore
| | - Joachim Hombach
- Initiative for Vaccine Research, World Health Organization, Geneva, Switzerland
| | - Immo Kleinschmidt
- Tropical Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Pathology, School of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Audrey Lenhart
- Centers for Disease Control and Prevention, Center for Global Health/Division of Parasitic Diseases and Malaria/Entomology Branch, Atlanta, Georgia, United States of America
| | - Steven W. Lindsay
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- School of Biological and Biomedical Sciences, Durham University, Durham, United Kingdom
| | - Ira Longini
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | | | - Amy C. Morrison
- Department of Entomology and Nematology, University of California, Davis, California, United States of America
| | - T. Alex Perkins
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Gonzalo Vazquez-Prokopec
- Department of Environmental Studies, Emory University, Atlanta, Georgia, United States of America
| | - Paul Reiter
- Department of Medical Entomology, Institut Pasteur, Paris, France
| | - Scott A. Ritchie
- College of Public Health, Medical, and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - David L. Smith
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, Washington, United States of America
| | - Daniel Strickman
- Bill & Melinda Gates Foundation, Seattle, Washington, United States of America
| | - 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, California, United States of America
| |
Collapse
|
736
|
Hladish TJ, Pearson CAB, Chao DL, Rojas DP, Recchia GL, Gómez-Dantés H, Halloran ME, Pulliam JRC, Longini IM. Projected Impact of Dengue Vaccination in Yucatán, Mexico. PLoS Negl Trop Dis 2016; 10:e0004661. [PMID: 27227883 PMCID: PMC4882069 DOI: 10.1371/journal.pntd.0004661] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 04/02/2016] [Indexed: 01/17/2023] Open
Abstract
Dengue vaccines will soon provide a new tool for reducing dengue disease, but the effectiveness of widespread vaccination campaigns has not yet been determined. We developed an agent-based dengue model representing movement of and transmission dynamics among people and mosquitoes in Yucatán, Mexico, and simulated various vaccine scenarios to evaluate effectiveness under those conditions. This model includes detailed spatial representation of the Yucatán population, including the location and movement of 1.8 million people between 375,000 households and 100,000 workplaces and schools. Where possible, we designed the model to use data sources with international coverage, to simplify re-parameterization for other regions. The simulation and analysis integrate 35 years of mild and severe case data (including dengue serotype when available), results of a seroprevalence survey, satellite imagery, and climatological, census, and economic data. To fit model parameters that are not directly informed by available data, such as disease reporting rates and dengue transmission parameters, we developed a parameter estimation toolkit called AbcSmc, which we have made publicly available. After fitting the simulation model to dengue case data, we forecasted transmission and assessed the relative effectiveness of several vaccination strategies over a 20 year period. Vaccine efficacy is based on phase III trial results for the Sanofi-Pasteur vaccine, Dengvaxia. We consider routine vaccination of 2, 9, or 16 year-olds, with and without a one-time catch-up campaign to age 30. Because the durability of Dengvaxia is not yet established, we consider hypothetical vaccines that confer either durable or waning immunity, and we evaluate the use of booster doses to counter waning. We find that plausible vaccination scenarios with a durable vaccine reduce annual dengue incidence by as much as 80% within five years. However, if vaccine efficacy wanes after administration, we find that there can be years with larger epidemics than would occur without any vaccination, and that vaccine booster doses are necessary to prevent this outcome. Dengue is a mosquito-transmitted viral disease that is common throughout the tropics. Despite a long history in humans and extensive efforts to control dengue transmission in many countries, the number, severity, and geographic range of reported cases is increasing. Most control efforts have focused on controlling mosquito populations, but the main vector, Aedes aegypti, flourishes in human-disturbed and indoor environments. Because the mosquitoes prefer to bite during the day when people are active and potentially moving around high-risk locations, fixed barriers like bed nets are not effective. Several dengue vaccines are being actively developed and may become valuable tools in dengue control. Using historical dengue data from Yucatán, Mexico, we fit a detailed simulation of human and mosquito populations to project future transmission, then used efficacy data from vaccine trials to evaluate the benefit of potential vaccination deployment strategies in the region. For a durable vaccine, we find that population-level, annual vaccine effectiveness approaches 65% by the end of the 20-year forecast period. For waning vaccines, however, effectiveness is greatly reduced–and sometimes negative–unless booster vaccinations are used.
Collapse
Affiliation(s)
- Thomas J. Hladish
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| | - Carl A. B. Pearson
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Dennis L. Chao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Diana Patricia Rojas
- Department of Epidemiology, University of Florida, Gainesville, Florida, United States of America
| | - Gabriel L. Recchia
- Institute for Intelligent Systems, University of Memphis, Memphis, Tennessee, United States of America
| | - Héctor Gómez-Dantés
- Health Systems Research Center, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - M. Elizabeth Halloran
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Center for Inference and Dynamics of Infectious Diseases, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Juliet R. C. Pulliam
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Ira M. Longini
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
737
|
Use of a Rapid Test for Diagnosis of Dengue during Suspected Dengue Outbreaks in Resource-Limited Regions. J Clin Microbiol 2016; 54:2090-5. [PMID: 27225409 DOI: 10.1128/jcm.00521-16] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/23/2016] [Indexed: 12/19/2022] Open
Abstract
Dengue is major public health problem, globally. Timely verification of suspected dengue outbreaks allows for public health response, leading to the initiation of appropriate clinical care. Because the clinical presentation of dengue is nonspecific, dengue diagnosis would benefit from a sensitive rapid diagnostic test (RDT). We evaluated the diagnostic performance of an RDT that detects dengue virus (DENV) nonstructural protein 1 (NS1) and anti-DENV IgM during suspected acute febrile illness (AFI) outbreaks in four countries. Real-time reverse transcription-PCR and anti-DENV IgM enzyme-linked immunosorbent assay were used to verify RDT results. Anti-DENV IgM RDT sensitivity and specificity ranged from 55.3 to 91.7% and 85.3 to 98.5%, respectively, and NS1 sensitivity and specificity ranged from 49.7 to 92.9% and 22.2 to 89.0%, respectively. Sensitivity varied by timing of specimen collection and DENV serotype. Combined test results moderately improved the sensitivity. The use of RDTs identified dengue as the cause of AFI outbreaks where reference diagnostic testing was limited or unavailable.
Collapse
|
738
|
Harapan H, Anwar S, Setiawan AM, Sasmono RT. Dengue vaccine acceptance and associated factors in Indonesia: A community-based cross-sectional survey in Aceh. Vaccine 2016; 34:3670-5. [PMID: 27208588 DOI: 10.1016/j.vaccine.2016.05.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND The first dengue vaccine (DV) has been licensed in some countries, but an assessment of the public's acceptance of DV is widely lacking. This study aimed to explore and understand DV acceptance and its associated explanatory variables among healthy inhabitants of Aceh, Indonesia. METHODS A community-based cross-sectional survey was conducted from November 2014 to March 2015 in nine regencies of Aceh that were selected randomly. A set of validated questionnaires covering a range of explanatory variables and DV acceptance was used to conduct the interviews. A multi-step logistic regression analysis and Spearman's rank correlation were employed to assess the role of explanatory variables in DV acceptance. RESULTS We included 652 community members in the final analysis and found that 77.3% of them were willing to accept the DV. Gender, monthly income, socioeconomic status (SES), attitude toward dengue fever (DF) and attitude toward vaccination practice were associated with DV acceptance in bivariate analyses (P<0.05). A correlation analysis confirmed that attitude toward vaccination practice and attitude toward DF were strongly correlated with DV acceptance, rs=0.41 and rs=0.39, respectively (P<0.001). The multivariate analysis revealed that a high monthly income, high SES, and a good attitude toward vaccination practice and toward DF were independent predictors of DV acceptance. CONCLUSION The acceptance rate of the DV among inhabitants of Aceh, Indonesia was relatively high, and the strongest associated factors of higher support for the DV were a good attitude toward vaccination practices and a good attitude toward DF.
Collapse
Affiliation(s)
- Harapan Harapan
- Medical Research Unit, School of Medicine, Syiah Kuala University, Banda Aceh, Indonesia; Tropical Disease Centre, School of Medicine, Syiah Kuala University, Banda Aceh, Indonesia; Department of Microbiology, School of Medicine, Syiah Kuala University, Banda Aceh, Indonesia.
| | - Samsul Anwar
- Department of Statistics, Faculty of Mathematics and Natural Sciences, Syiah Kuala University, Banda Aceh, Indonesia
| | - Abdul Malik Setiawan
- Faculty of Medicine and Health Sciences, State Islamic University Maulana Malik Ibrahim, Malang, Indonesia
| | | | | |
Collapse
|
739
|
Lauretti F, Chattopadhyay A, de Oliveira França RF, Castro-Jorge L, Rose J, Fonseca BALD. Recombinant vesicular stomatitis virus-based dengue-2 vaccine candidate induces humoral response and protects mice against lethal infection. Hum Vaccin Immunother 2016; 12:2327-33. [PMID: 27185081 DOI: 10.1080/21645515.2016.1183857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dengue is the most important arbovirus disease throughout the world and it is responsible for more than 500,000 dengue hemorrhagic cases and 22,000 deaths every year. One vaccine was recently licensed for human use in Brazil, Mexico and Philippines and although at least seven candidates have been in clinical trials the results of the most developed CYD vaccine have demonstrated immunization problems, such as uneven protection and interference between serotypes. We constructed a vaccine candidate based on vesicular stomatitis virus (VSV) expression of pre-membrane (prM) and envelope (E) proteins of dengue-2 virus (DENV-2) and tested it in mice to evaluate immunogenicity and protection against DENV-2 infection. VSV has been successfully used as vaccine vectors for several viruses to induce strong humoral and cellular immune responses. The VSV-DENV-2 recombinant was constructed by inserting the DENV-2 structural proteins into a VSV plasmid DNA for recombinant VSV-DENV-2 recovery. Infectious recombinant VSV viruses were plaque purified and prM and E expression were confirmed by immunofluorescence and radiolabeling of proteins of infected cells. Forty Balb/C mice were inoculated through subcutaneous (s.c.) route with VSV-DENV-2 vaccine in a two doses schedule 15 d apart and 29 d after first inoculation, sera were collected and the mice were challenged with 50 lethal doses (LD50) of a neurovirulent DENV-2. The VSV-DENV-2 induced anti-DENV-2 antibodies and protected animals in the challenge experiment comparable to DENV-2 immunization control group. We conclude that VSV is a promising platform to test as a DENV vaccine and perhaps against others Flaviviridae.
Collapse
Affiliation(s)
- Flavio Lauretti
- a Department of Internal Medicine , School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto - SP , Brazil
| | - Anasuya Chattopadhyay
- b Department of Pathology , Yale University School of Medicine , New Haven , CT , USA
| | | | - Luiza Castro-Jorge
- a Department of Internal Medicine , School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto - SP , Brazil
| | - John Rose
- b Department of Pathology , Yale University School of Medicine , New Haven , CT , USA
| | - Benedito A L da Fonseca
- a Department of Internal Medicine , School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto - SP , Brazil
| |
Collapse
|
740
|
Ye Q, Liu ZY, Han JF, Jiang T, Li XF, Qin CF. Genomic characterization and phylogenetic analysis of Zika virus circulating in the Americas. INFECTION GENETICS AND EVOLUTION 2016; 43:43-9. [PMID: 27156653 DOI: 10.1016/j.meegid.2016.05.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 02/04/2023]
Abstract
The rapid spread and potential link with birth defects have made Zika virus (ZIKV) a global public health problem. The virus was discovered 70years ago, yet the knowledge about its genomic structure and the genetic variations associated with current ZIKV explosive epidemics remains not fully understood. In this review, the genome organization, especially conserved terminal structures of ZIKV genome were characterized and compared with other mosquito-borne flaviviruses. It is suggested that major viral proteins of ZIKV share high structural and functional similarity with other known flaviviruses as shown by sequence comparison and prediction of functional motifs in viral proteins. Phylogenetic analysis demonstrated that all ZIKV strains circulating in the America form a unique clade within the Asian lineage. Furthermore, we identified a series of conserved amino acid residues that differentiate the Asian strains including the current circulating American strains from the ancient African strains. Overall, our findings provide an overview of ZIKV genome characterization and evolutionary dynamics in the Americas and point out critical clues for future virological and epidemiological studies.
Collapse
Affiliation(s)
- Qing Ye
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Zhong-Yu Liu
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Jian-Feng Han
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Tao Jiang
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Xiao-Feng Li
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China
| | - Cheng-Feng Qin
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, China.
| |
Collapse
|
741
|
[Zika virus infection or the future of infectious diseases]. Med Clin (Barc) 2016; 147:300-5. [PMID: 27156484 PMCID: PMC7094702 DOI: 10.1016/j.medcli.2016.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/02/2022]
Abstract
El virus Zika es un Flavivirus filogenéticamente cercano al de la fiebre amarilla o del dengue, cuyo vector principal es el mosquito Aedes aegypti. El virus procede de un reservorio simiano africano y ha protagonizado una expansión fulminante a través del Pacífico hasta Sudamérica. Provoca una enfermedad leve caracterizada por fiebre con exantema. La mortalidad se circunscribe a casos de Guillain-Barré y de malformación encefálica fetal con microcefalia. Un caso sospechoso será aquel con: a) antecedente epidemiológico de desplazamiento a zona endémica; b) cuadro pseudogripal con exantema, y c) hemograma/bioquímica levemente alteradas o normales. La confirmación diagnóstica requiere identificar al virus por RT-PCR en sangre (hasta el quinto día sintomático), orina (hasta el día 10-14) o IgM específicas a partir del quinto día. Existe alguna evidencia que da soporte a la relación causa-efecto con la microcefalia fetal. A la espera de datos definitivos, las mujeres embarazadas procedentes de Centro y Sudamérica deben ser testadas para descartar la infección.
Collapse
|
742
|
Introducing dengue vaccine: Implications for diagnosis in dengue vaccinated subjects. Vaccine 2016; 34:2759-61. [PMID: 27142330 DOI: 10.1016/j.vaccine.2016.04.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 11/24/2022]
Abstract
Diagnosis of dengue virus infections is complicated by preference for different diagnostic tests in different post onset days of illness and the presence of multiple serotypes leading to secondary and tertiary infections. The sensitivity of the most commonly employed diagnostic assays such as anti dengue IgM capture (MAC) ELISA and non structural protein (NS) 1 capture ELISA are lower in secondary and subsequent infections. Introduction of dengue vaccine in endemic regions will affect the way how dengue is diagnosed in vaccinated subjects. This viewpoint article discusses implications of introduction of dengue vaccine on the diagnosis of dengue infections in vaccinated subjects and the strategies that are needed to tackle the issue.
Collapse
|
743
|
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
|
744
|
|
745
|
Gessner BD, Wilder-Smith A. Estimating the public health importance of the CYD-tetravalent dengue vaccine: Vaccine preventable disease incidence and numbers needed to vaccinate. Vaccine 2016; 34:2397-401. [PMID: 27055020 DOI: 10.1016/j.vaccine.2016.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 03/01/2016] [Accepted: 03/09/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND To evaluate the potential public health impact of the live attenuated tetravalent Sanofi Pasteur dengue vaccine (CYD-TDV) we analyzed data from the reported clinical trials to calculate vaccine preventable disease incidence (VPDI) and number needed to vaccinate (NNV) based on the licensure indication for persons age 9 years and above. METHODS VPDI is defined as incidence in an unvaccinated population X vaccine efficacy (VE), and thus incorporates both VE and the underlying burden of disease. NNV was calculated as 100,000 divided by VPDI divided by 2-year length of study. We compared these values to data for three newer vaccines that are currently integrated into some national immunization programs in Asia and Latin America, namely pneumococcal conjugate, Haemophilus influenzae type b, and rotavirus vaccines. RESULTS In the Asian-Pacific trial, in the first 25 months after the first dose of the dengue vaccine, CYD-TDV prevented annually 2639 cases of virologically confirmed dengue for every 100,000 persons vaccinated, for an NNV of 18. In the Latin American trial, given the overall lower annual dengue incidence compared to Asia, VPDI was 1707, and NNV 28. For the Asian-Pacific and Latin American studies, the VPDIs for hospitalized virologically confirmed disease at the trials' end were 638 and 239 per 100,000 population per year, respectively, with NNVs of 75 and 201. VPDI for confirmed dengue hospitalization was higher than that for Hib vaccine against Hib meningitis or all cause severe pneumonia while lower than that for rotavirus vaccine against severe rotavirus gastroenteritis. CONCLUSIONS Our analysis found that the CYD-TDV dengue vaccine had favorable VPDI and NNV, also when compared to existing vaccines used in Latin America and Asia. VPDI and NNV varied by serotype distribution, extent of prior dengue exposure (baseline seroprevalence) and country. These findings will help policy-makers decide where and how to introduce this vaccine post-licensure.
Collapse
Affiliation(s)
| | - Annelies Wilder-Smith
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Institute of Public Health, University of Heidelberg, Germany.
| |
Collapse
|
746
|
Fan Y, Moon JJ. Particulate delivery systems for vaccination against bioterrorism agents and emerging infectious pathogens. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27038091 DOI: 10.1002/wnan.1403] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/10/2016] [Accepted: 02/15/2016] [Indexed: 01/15/2023]
Abstract
Bioterrorism agents that can be easily transmitted with high mortality rates and cause debilitating diseases pose major threats to national security and public health. The recent Ebola virus outbreak in West Africa and ongoing Zika virus outbreak in Brazil, now spreading throughout Latin America, are case examples of emerging infectious pathogens that have incited widespread fear and economic and social disruption on a global scale. Prophylactic vaccines would provide effective countermeasures against infectious pathogens and biological warfare agents. However, traditional approaches relying on attenuated or inactivated vaccines have been hampered by their unacceptable levels of reactogenicity and safety issues, whereas subunit antigen-based vaccines suffer from suboptimal immunogenicity and efficacy. In contrast, particulate vaccine delivery systems offer key advantages, including efficient and stable delivery of subunit antigens, co-delivery of adjuvant molecules to bolster immune responses, low reactogenicity due to the use of biocompatible biomaterials, and robust efficiency to elicit humoral and cellular immunity in systemic and mucosal tissues. Thus, vaccine nanoparticles and microparticles are promising platforms for clinical development of biodefense vaccines. In this review, we summarize the current status of research efforts to develop particulate vaccine delivery systems against bioterrorism agents and emerging infectious pathogens. WIREs Nanomed Nanobiotechnol 2017, 9:e1403. doi: 10.1002/wnan.1403 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Yuchen Fan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
747
|
Adde A, Roucou P, Mangeas M, Ardillon V, Desenclos JC, Rousset D, Girod R, Briolant S, Quenel P, Flamand C. Predicting Dengue Fever Outbreaks in French Guiana Using Climate Indicators. PLoS Negl Trop Dis 2016; 10:e0004681. [PMID: 27128312 PMCID: PMC4851397 DOI: 10.1371/journal.pntd.0004681] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/11/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Dengue fever epidemic dynamics are driven by complex interactions between hosts, vectors and viruses. Associations between climate and dengue have been studied around the world, but the results have shown that the impact of the climate can vary widely from one study site to another. In French Guiana, climate-based models are not available to assist in developing an early warning system. This study aims to evaluate the potential of using oceanic and atmospheric conditions to help predict dengue fever outbreaks in French Guiana. METHODOLOGY/PRINCIPAL FINDINGS Lagged correlations and composite analyses were performed to identify the climatic conditions that characterized a typical epidemic year and to define the best indices for predicting dengue fever outbreaks during the period 1991-2013. A logistic regression was then performed to build a forecast model. We demonstrate that a model based on summer Equatorial Pacific Ocean sea surface temperatures and Azores High sea-level pressure had predictive value and was able to predict 80% of the outbreaks while incorrectly predicting only 15% of the non-epidemic years. Predictions for 2014-2015 were consistent with the observed non-epidemic conditions, and an outbreak in early 2016 was predicted. CONCLUSIONS/SIGNIFICANCE These findings indicate that outbreak resurgence can be modeled using a simple combination of climate indicators. This might be useful for anticipating public health actions to mitigate the effects of major outbreaks, particularly in areas where resources are limited and medical infrastructures are generally insufficient.
Collapse
Affiliation(s)
- Antoine Adde
- Unité d’épidémiologie, Institut Pasteur de la Guyane, Cayenne, Guyane
- Unité d’entomologie médicale, Institut Pasteur de la Guyane, Cayenne, Guyane
| | - Pascal Roucou
- Centre de Recherches de Climatologie, UMR6282 Biogéosciences, CNRS Université de Bourgogne Franche-Comté, Dijon, France
| | - Morgan Mangeas
- Maison de la Télédétection, UMR 228 ESPACE-DEV, Institut de Recherche pour le Développement, Montpellier, France
| | - Vanessa Ardillon
- Cellule de l’Institut de Veille Sanitaire en Régions Antilles - Guyane, Cayenne, Guyane
| | | | | | - Romain Girod
- Unité d’entomologie médicale, Institut Pasteur de la Guyane, Cayenne, Guyane
| | - Sébastien Briolant
- Unité d’entomologie médicale, Institut Pasteur de la Guyane, Cayenne, Guyane
- Direction Interarmées du Service de Santé en Guyane, Cayenne, Guyane
- Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
| | - Philippe Quenel
- Laboratoire d’Etudes et de Recherche en Santé-Environnement, Ecole des Hautes Etudes en Santé Publique, Rennes, France
| | - Claude Flamand
- Unité d’épidémiologie, Institut Pasteur de la Guyane, Cayenne, Guyane
| |
Collapse
|
748
|
Poland GA, Whitaker JA, Poland CM, Ovsyannikova IG, Kennedy RB. Vaccinology in the third millennium: scientific and social challenges. Curr Opin Virol 2016; 17:116-125. [PMID: 27039875 PMCID: PMC4902778 DOI: 10.1016/j.coviro.2016.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 02/19/2016] [Accepted: 03/11/2016] [Indexed: 12/21/2022]
Abstract
The epidemiology of deaths due to vaccine-preventable diseases has been significantly and positively altered through the use of vaccines. Despite this, significant challenges remain in vaccine development and use in the third millennium. Both new (Ebola, Chikungunya, Zika, and West Nile) and re-emerging diseases (measles, mumps, and influenza) require the development of new or next-generation vaccines. The global aging of the population, and accumulating numbers of immunocompromised persons, will require new vaccine and adjuvant development to protect large segments of the population. After vaccine development, significant challenges remain globally in the cost and efficient use and acceptance of vaccines by the public. This article raises issues in these two areas and suggests a way forward that will benefit current and future generations.
Collapse
Affiliation(s)
- Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Jennifer A Whitaker
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Caroline M Poland
- Taylor University Counseling Center, Taylor University, Upland, IN 46989, USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
749
|
Forshey BM, Stoddard ST, Morrison AC. Dengue Viruses and Lifelong Immunity: Reevaluating the Conventional Wisdom. J Infect Dis 2016; 214:979-81. [PMID: 26984147 DOI: 10.1093/infdis/jiw102] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 11/12/2022] Open
Affiliation(s)
- Brett M Forshey
- Global Emerging Infections Surveillance and Response Section, Armed Forces Health Surveillance Branch Cherokee Nation Technology Solutions, Silver Spring, Maryland
| | | | - Amy C Morrison
- Department of Entomology and Nematology, University of California, Davis
| |
Collapse
|
750
|
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: 194] [Impact Index Per Article: 21.6] [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
|