1,550
Views
34
CrossRef citations to date
0
Altmetric
Special Focus Commentary

Dengue human infection model

Re-establishing a tool for understanding dengue immunology and advancing vaccine development

Pages 1587-1590 | Received 20 Feb 2013, Accepted 04 Mar 2013, Published online: 06 Mar 2013

Abstract

Dengue is an emerging and re-emerging disease of the tropics and sub-tropics. Millions of infections occur annually exacting a significant social, financial, and health care resource toll. Widespread use of a safe and efficacious dengue vaccine in cooperation with strategic vector control is the best hope for reducing the global dengue burden. Despite over 100 y of research exploring dengue immunology, pathogenesis, animal models, and vaccine and drug development there is no licensed vaccine or dengue anti-viral. No correlate of protection or validated animal model of disease has been defined. Experimental human infection with partially attenuated dengue viruses are documented as early as 1902 and have facilitated research efforts resulting in seminal discoveries and observations. It is time to explore re-invigorating the dengue human infection model to support dengue vaccine development.

The world needs a dengue vaccine. Unfortunately, diverse efforts and approaches spanning more than 70 y have failed to achieve this goal.Citation1 As a neglected tropical disease, dengue and the dengue vaccine field have not received the consistent financial or political support benefitting other infectious diseases. However, fundamental scientific challenges have also contributed to slowing vaccine development efforts. It is time to explore re-establishing the dengue human infection model (DHIM) to support dengue medical countermeasure development and advance the field’s understanding of this disease’s unique immuno-pathogenesis.

Dengue is the world’s most rapidly expanding and important arboviral disease.Citation2 Millions of people are infected, millions suffer symptomatic disease requiring medical care, and tens of thousands die every year from endemic and epidemic transmission.Citation3(WHO, 2013. Dengue and severe dengue, Fact sheet N°117. Available at: www.who.int/mediacentre/factsheets/fs117/en/index.html, accessed February, 2013.) Dengue is a leading cause of febrile illness in the returning traveler and has impacted military operations since World War II.Citation4-Citation13 Although mortality is relatively low compared with other infectious diseases of the developing world, dengue exacts a significant social, financial, and health care resource toll.Citation14-Citation17

There is no licensed vaccine or therapeutic to prevent or treat DENV infection or disease. As such, many endemic countries execute costly prophylactic or reactionary vector control measures, often without noticeable impact. Population growth and urbanization, changing ecologic conditions favoring expanding Aedes mosquito habitats, and the increasing ease and access to regional and international travel will continue to drive the worsening global dengue burden.Citation18

In 2012 there was expectant optimism surrounding the results of the world’s first dengue vaccine efficacy trial. Optimism gave way to confusion and disappointment as the vaccine’s sponsor published in The Lancet an overall per-protocol efficacy of 30.2%. The trial occurred in a population of Thai children highly immunologically primed to Japanese encephalitis virus and/or dengue, a condition many thought would improve the vaccine’s performance. Efficacy across the dengue virus (DENV) types was unequal; efficacy against disease following DENV-2 infection was the lowest (9.2%). Neutralizing antibody responses in vaccine recipients compared with control were robust and balanced. There was no evidence symptomatic breakthrough infections in the vaccine group were more severe than in controls. Numerous hypotheses have been proposed to explain the study findings and the apparent disconnect between immunogenicity and clinical efficacy. Unfortunately, the size and blood sample collection schedule of the per-protocol immunogenicity volunteer subset is unlikely to support a comprehensive retrospective forensic analysis.Citation19

The limited success of this first efficacy trial has highlighted a number of questions for the field:

  • Is the importance of neutralizing antibody in question as it relates to its role in the protective immune response following natural infection or it’s potential to be validated as an immune correlate of protection?

  • Is development of DENV type-specific cellular immune responses against the DENV non-structural proteins a requirement for protection?

  • Have concerns increased regarding the limitations of the plaque reduction neutralization test (PRNT) assay platform and its ability to measure vaccine immunogenicity?

  • Do dengue vaccine developers need to increase their consideration of DENV diversity and evolution when developing vaccine candidates?

  • Is it possible population-level genetic background and pre-existing antibody levels to dengue and/or non-dengue flaviviruses could significantly impact vaccine candidate performance?

One method of answering these questions would be a prospective cohort study measuring clinical endpoints. Scheduled cohort blood sample collections would allow retrospective analyses of the relationships between interesting clinical endpoints (e.g., any dengue, any clinical severity, caused by any infecting DENV type) and immune profiles (i.e., neutralizing antibody titer) measured in pre-illness samples. These studies are expensive and require enrollment of large sample sizes to address clinical attack rates of 1–2% per year.Citation20,Citation21 Risk of capturing sufficient clinical disease is risky in areas with focal and unpredictable transmission dynamics.Citation22,Citation23 The more narrow the study questions (i.e., study of severe dengue, study of disease caused by a specific DENV type, etc.) the larger the study.

Another option for exploring the relationship between immune profiles and protection would be to use a well-characterized dengue disease animal model. Although significant progress has been made in this area, gaps remain and vaccine developers are reluctant to base important development decisions (i.e., investing in a clinical endpoint study) on a candidate’s efficacy demonstrated in a mouse model.Citation24 Non-human primates (NHP) develop viremia and a neutralizing antibody response following exposure but usually do not become ill.Citation25,Citation26 Therefore, vaccine development decisions are guided by extrapolating efficacy data from NHP studies (i.e., prevention of viremia in vaccinated vs. controls following challenge) and assumptions about the association between neutralizing antibody profiles (i.e., magnitude, balance) observed in phase 1 and 2 clinical studies and the candidate’s potential for clinical benefit.Citation27

Response to The Lancet publication should be measured and final judgment on vaccine performance reserved until phase 3 data from a broader population and geographic area is available. It is prudent, however, for dengue vaccine developers to believe their development programs may be at increased risk if efficacy in a NHP dengue infection model and neutralizing antibody are not predictive or associated with human clinical efficacy in any meaningful way or in any way which can be measured. For these reasons, it is time to explore re-establishing a DHIM.

For decades, controlled human infection (challenge) studies have been used to develop vaccine candidates and study infectious disease immunology.Citation28-Citation33 The ethics of intentionally infecting healthy volunteers has been vigorously debated and much has been written on the topic.Citation34-Citation36 A discussion of these issues is beyond the scope of these comments. However, one unique challenge of a DHIM, which many believe differentiates it from other controlled infection scenarios, is the absence of any specific anti-dengue therapeutic (i.e., anti-viral). Although specific treatment algorithms do exist and in experienced hands work very well to prevent morbidity and mortality, many find the absence of a specific anti-dengue therapeutic concerning. DHIM developers and end-users will be required by regulatory agencies and institutional review boards to calculate and articulate the risk of a bad outcome (i.e., severe dengue, dengue hemorrhagic fever) following infection with a slightly attenuated DENV. In-depth descriptions of measures to reduce volunteer risk and clinical management guidelines will need to accompany protocol submission packets.

Descriptions of controlled dengue human infection experiments can be found in the literature as early as 1902 and as recent as 2013.Citation1,Citation37-Citation47 According to a literature review, more than 670 volunteers have been experimentally infected using both needle and mosquito virus delivery. Early foundational understandings about dengue were produced using human infection experiments. Discoveries and observations included: the viral etiology of dengue, virus transmission mechanisms, viral incubation period in mosquito and man, period of infectivity in mosquito and man, competence of Aedes species to transmit virus and the incompetence of Culex species, clinical and clinical laboratory features of uncomplicated dengue illness, development of homologous immunity following infection, the existence of multiple DENV types, demonstration of anti-dengue virus neutralizing antibodies, and the utility of a DHIM in assessing vaccine candidates. More recent studies have reported ultrasonographic findings, characteristics of cellular immune responses, and clinical and clinical laboratory findings following experimental infection of flavivirus naïve and previously vaccinated (experimental candidate) individuals.Citation48-Citation50

A well-characterized DHIM would be a valuable research tool. Vaccine and drug development pathways, immunologic investigations of pathogenesis, exploring immune correlates of protection, and augmenting regulatory strategies for licensing and labeling products would all benefit. Vaccinologists, drug developers, and immunologists discussed these concepts at a Walter Reed Army Institute of Research (WRAIR) and National Institutes of Health (NIH) co-sponsored, “Dengue Human Infection Model,” workshop in 2011 and a recent Wellcome Trust event entitled, “Controlled Human Infection Studies in the Development of Vaccines and Therapeutics.” Comments here will focus on vaccine development.

A DHIM could support various elements of the vaccine clinical development life cycle. Vaccine developers and scientists could test their candidate’s safety and potential for clinical efficacy across a number of conditions and scenarios. Endemic countries introducing a dengue vaccine into their national immunization program will undoubtedly vaccinate numerous previously infected individuals. Therefore, a dengue vaccine will need to establish, early in development, safety in dengue primed individuals.Citation51 Initial explorations of safety in dengue primed individuals typically require execution of phase 1 or 2 studies in dengue endemic countries.Citation52,Citation53 Preparing for and conducting these studies is expensive and painstakingly slow. Depending on the region, study volunteers may also be primed to other, non-dengue flaviviruses (i.e., Yellow fever, Japanese encephalitis) making data interpretation difficult. A DHIM would allow developers to create dengue primed individuals and complete preliminary safety testing on a smaller scale and in very controlled and lower risk environments. The impact of dengue priming on safety as a function of time (i.e., recent vs. remote priming) could also be evaluated. Priming to different DENV types would be possible. In-depth clinical and immunologic characterizations of volunteer reactions to vaccination could generate early safety benchmarks and promote hypotheses for observed outcomes (i.e., reactogenicity associated with pro-inflammatory cellular immune responses).

A DHIM could also support early vaccine formulation decisions including antigen selection, antigen concentration, balancing antigen input, and adjuvant selection and dosing. Challenging vaccine recipients across different treatment groups and candidate formulations would supplement NHP and human immunogenicity data to allow more informed decision-making earlier in development. If a DHIM for every DENV type was available, developers could probe DENV-specific vaccine performance and target re-formulation or re-derivation efforts. Improving vaccine performance or other aspects of the target product profile (TPP) with an adjuvant could also be explored.

One of the most valuable uses of a DHIM would be supporting the discovery of an immune correlate and surrogate of protection. Correlating pre-infection immune profiles with post-experimental infection clinical outcomes would allow scientists to explore how humoral and/or cellular immunity readouts may be associated with protection. It is possible vaccine-induced neutralizing antibodies or interferon gamma responses may be associated with disease attenuation or prevention (i.e., correlate). Quantitative protective thresholds or breakpoints for individual endpoints may also exist (i.e., surrogate). Correlates and surrogates would be viewed in the context of the specific assay platform and executing laboratory (i.e., vaccine developer “X” using a microneutralization platform) and determined in a DENV type-specific manner. The timeframe between vaccination and sample collection (i.e., 1 mo post-completion of the vaccine regimen, 6 mo post-completion, etc.) would be considered. DHIM-derived correlates and surrogates would allow developers to establish target benchmarks for vaccine candidate performance in early phase 1 and 2 studies. Candidates not meeting the threshold could be down-selected or re-derived. Recognizing surrogate threshold may not hold true following infection with an un-attenuated wild type DENV captured during an efficacy trial (i.e., DHIM threshold too low), DHIM derived correlates and surrogates would need to be validated by associating them with actual efficacy and immunogenicity endpoints measured during a field trial.

The above represents an incomplete list of potential uses for a DHIM in dengue vaccine development. Uses related to exploring basic immunology, clinical pathology, drug development, and diagnostics also exist. Augmenting regulatory strategy for licensing vaccines or drugs in non-endemic countries requires further discussion with national regulatory authorities.

The DHIM would bring value to vaccine development efforts because of its high “negative predictive value.” A vaccine candidate capable of protecting a volunteer following infection with a slightly attenuated DHIM viral strain may be protective in a field efficacy study with natural wild-type viruses. On the other hand, there is little chance a vaccine candidate would be protective in the field if it was not protective in the context of a DHIM study. The need for a dengue vaccine is too great and efforts to develop a vaccine have proceeded unsuccessfully for too long to not explore a DHIM’s utility and potential concerns.

Abbreviations:
DENV=

dengue virus

DHIM=

dengue human infection model

NIH=

National Institutes of Health

NHP=

non-human primate

PRNT=

plaque reduction neutralization test

TPP=

target product profile

WRAIR=

Walter Reed Army Institute of Research

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Disclaimer

The opinions or assertions contained herein are the private views of the author and are not to be construed as reflecting the official views of the United States Army or the United States Department of Defense.

References

  • Simmons JS, St John JH, Reynolds FHK. Experimental studies of dengue. Philipp J Sci 1931; 44:1 - 252
  • Guzman MG, Halstead SB, Artsob H, Buchy P, Farrar J, Gubler DJ, et al. Dengue: a continuing global threat. Nat Rev Microbiol 2010; 8:Suppl S7 - 16; http://dx.doi.org/10.1038/nrmicro2460; PMID: 21079655
  • Beatty ME, Stone A, Fitzsimons DW, Hanna JN, Lam SK, Vong S, et al, Asia-Pacific and Americas Dengue Prevention Boards Surveillance Working Group. Best practices in dengue surveillance: a report from the Asia-Pacific and Americas Dengue Prevention Boards. PLoS Negl Trop Dis 2010; 4:e890; http://dx.doi.org/10.1371/journal.pntd.0000890; PMID: 21103381
  • Centers for Disease Control and Prevention (CDC). Travel-associated Dengue surveillance - United States, 2006-2008. MMWR Morb Mortal Wkly Rep 2010; 59:715 - 9; PMID: 20559202
  • Chen LH, Wilson ME. Dengue and chikungunya in travelers: recent updates. Curr Opin Infect Dis 2012; 25:523 - 9; http://dx.doi.org/10.1097/QCO.0b013e328356ffd5; PMID: 22825287
  • Gibbons RV, Streitz M, Babina T, Fried JR. Dengue and US military operations from the Spanish-American War through today. Emerg Infect Dis 2012; 18:623 - 30; http://dx.doi.org/10.3201/eid1804.110134; PMID: 22469290
  • Hynes NA. Dengue: A reemerging concern for travelers. Cleve Clin J Med 2012; 79:474 - 82; http://dx.doi.org/10.3949/ccjm.79a.11048; PMID: 22751631
  • de Laval F, Plumet S, Simon F, Deparis X, Leparc-Goffart I. Dengue surveillance among French military in Africa. Emerg Infect Dis 2012; 18:342 - 3; http://dx.doi.org/10.3201/eid1802.111333; PMID: 22305127
  • Anna MM, Escobar JD, Chapman AS. Reported vectorborne and zoonotic diseases, U.S. Air Force, 2000-2011. MSMR 2012; 19:11 - 2, discussion 12-4; PMID: 23121007
  • Trofa AF, DeFraites RF, Smoak BL, Kanesa-thasan N, King AD, Burrous JM, et al. Dengue fever in US military personnel in Haiti. JAMA 1997; 277:1546 - 8; http://dx.doi.org/10.1001/jama.1997.03540430058033; PMID: 9153369
  • Sharp TW, Wallace MR, Hayes CG, Sanchez JL, DeFraites RF, Arthur RR, et al. Dengue fever in U.S. troops during Operation Restore Hope, Somalia, 1992-1993. Am J Trop Med Hyg 1995; 53:89 - 94; PMID: 7625541
  • Hayes CG, O’Rourke TF, Fogelman V, Leavengood DD, Crow G, Albersmeyer MM. Dengue fever in American military personnel in the Philippines: clinical observations on hospitalized patients during a 1984 epidemic. Southeast Asian J Trop Med Public Health 1989; 20:1 - 8; PMID: 2772694
  • Deller JJ Jr., Russell PK. An analysis of fevers of unknown origin in American soldiers in Vietnam. Ann Intern Med 1967; 66:1129 - 43; PMID: 5338939
  • Halasa YA, Shepard DS, Zeng W. Economic cost of dengue in Puerto Rico. Am J Trop Med Hyg 2012; 86:745 - 52; http://dx.doi.org/10.4269/ajtmh.2012.11-0784; PMID: 22556069
  • Gubler DJ. The economic burden of dengue. Am J Trop Med Hyg 2012; 86:743 - 4; http://dx.doi.org/10.4269/ajtmh.2012.12-0157; PMID: 22556068
  • Torres JR, Castro J. The health and economic impact of dengue in Latin America. Cadernos de saude publica / Ministerio da Saude, Fundacao Oswaldo Cruz. Escola Nacional de Saude Publica 2007; 23:Suppl 1 S23 - 31
  • Anderson KB, Chunsuttiwat S, Nisalak A, Mammen MP, Libraty DH, Rothman AL, et al. Burden of symptomatic dengue infection in children at primary school in Thailand: a prospective study. Lancet 2007; 369:1452 - 9; http://dx.doi.org/10.1016/S0140-6736(07)60671-0; PMID: 17467515
  • Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century. Trends Microbiol 2002; 10:100 - 3; http://dx.doi.org/10.1016/S0966-842X(01)02288-0; PMID: 11827812
  • Sabchareon A, Wallace D, Sirivichayakul C, Limkittikul K, Chanthavanich P, Suvannadabba S, et al. Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. Lancet 2012; 380:1559 - 67; http://dx.doi.org/10.1016/S0140-6736(12)61428-7; PMID: 22975340
  • Endy TP, Chunsuttiwat S, Nisalak A, Libraty DH, Green S, Rothman AL, et al. Epidemiology of inapparent and symptomatic acute dengue virus infection: a prospective study of primary school children in Kamphaeng Phet, Thailand. Am J Epidemiol 2002; 156:40 - 51; http://dx.doi.org/10.1093/aje/kwf005; PMID: 12076887
  • Endy TP, Anderson KB, Nisalak A, Yoon IK, Green S, Rothman AL, et al. Determinants of inapparent and symptomatic dengue infection in a prospective study of primary school children in Kamphaeng Phet, Thailand. PLoS Negl Trop Dis 2011; 5:e975; http://dx.doi.org/10.1371/journal.pntd.0000975; PMID: 21390158
  • Jarman RG, Holmes EC, Rodpradit P, Klungthong C, Gibbons RV, Nisalak A, et al. Microevolution of Dengue viruses circulating among primary school children in Kamphaeng Phet, Thailand. J Virol 2008; 82:5494 - 500; http://dx.doi.org/10.1128/JVI.02728-07; PMID: 18367520
  • Rico-Hesse R. Microevolution and virulence of dengue viruses. Adv Virus Res 2003; 59:315 - 41; http://dx.doi.org/10.1016/S0065-3527(03)59009-1; PMID: 14696333
  • Cassetti MC, Durbin A, Harris E, Rico-Hesse R, Roehrig J, Rothman A, et al. Report of an NIAID workshop on dengue animal models. Vaccine 2010; 28:4229 - 34; http://dx.doi.org/10.1016/j.vaccine.2010.04.045; PMID: 20434551
  • Zompi S, Harris E. Animal models of dengue virus infection. Viruses 2012; 4:62 - 82; http://dx.doi.org/10.3390/v4010062; PMID: 22355452
  • Sun W, Endy TP, Nisalak A, Gettayacamin M, Eckels KH, Putnak R, et al. Tetravalent live-attenuated dengue virus vaccination protects rhesus monkeys against viremia from virulent dengue virus challenge. American Society for Virology Annual Meeting. Fort Collins, CO, 2000.
  • Osorio JE, Brewoo JN, Silengo SJ, Arguello J, Moldovan IR, Tary-Lehmann M, et al. Efficacy of a tetravalent chimeric dengue vaccine (DENVax) in Cynomolgus macaques. Am J Trop Med Hyg 2011; 84:978 - 87; http://dx.doi.org/10.4269/ajtmh.2011.10-0592; PMID: 21633037
  • Sauerwein RW, Roestenberg M, Moorthy VS. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat Rev Immunol 2011; 11:57 - 64; http://dx.doi.org/10.1038/nri2902; PMID: 21179119
  • Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, et al. Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med 2011; 183:734 - 42; http://dx.doi.org/10.1164/rccm.201006-0833OC; PMID: 20889904
  • Janowicz DM, Ofner S, Katz BP, Spinola SM. Experimental infection of human volunteers with Haemophilus ducreyi: fifteen years of clinical data and experience. J Infect Dis 2009; 199:1671 - 9; http://dx.doi.org/10.1086/598966; PMID: 19432549
  • Atmar RL, Opekun AR, Gilger MA, Estes MK, Crawford SE, Neill FH, et al. Norwalk virus shedding after experimental human infection. Emerg Infect Dis 2008; 14:1553 - 7; http://dx.doi.org/10.3201/eid1410.080117; PMID: 18826818
  • Landmann JK, Prociv P. Experimental human infection with the dog hookworm, Ancylostoma caninum. Med J Aust 2003; 178:69 - 71; PMID: 12526725
  • Freedman DO. Experimental infection of human subject with Strongyloides species. Rev Infect Dis 1991; 13:1221 - 6; http://dx.doi.org/10.1093/clinids/13.6.1221; PMID: 1775856
  • Miller FG, Grady C. The ethical challenge of infection-inducing challenge experiments. Clin Infect Dis 2001; 33:1028 - 33; http://dx.doi.org/10.1086/322664; PMID: 11528576
  • Hope T, McMillan J. Challenge studies of human volunteers: ethical issues. J Med Ethics 2004; 30:110 - 6; http://dx.doi.org/10.1136/jme.2003.004440; PMID: 14872087
  • Killingley B, Enstone J, Booy R, Hayward A, Oxford J, Ferguson N, et al, influenza transmission strategy development group. Potential role of human challenge studies for investigation of influenza transmission. Lancet Infect Dis 2011; 11:879 - 86; http://dx.doi.org/10.1016/S1473-3099(11)70142-6; PMID: 21798808
  • Graham H. Dengue: a study of its mode of propagation and pathology. MedRecNew york 1902; 61:204-7.
  • Graham H. The dengue: a study of its pathology and mode of propagation. J Trop Med 1903;
  • Ashburn PM, Craig CF. Experimental investigations regarding the etiology of dengue fever. J Infect Dis 1907; 4:440 - 75; http://dx.doi.org/10.1093/infdis/4.3.440; PMID: 15116315
  • Siler JF, Hall MW, Hitchens AP. Dengue: Its history, epidemilogy, mechanism of transmission, etiology, clinical manifestations, immunity, and prevention. Philipp J Sci 1926; 29:1 - 304
  • Cleland JB, Bradley B, Macdonald W. Further experiments in the etiology of dengue fever. J Hyg (Lond) 1919; 18:217 - 54; http://dx.doi.org/10.1017/S0022172400007476; PMID: 20474685
  • Sawada T, Sato H, Sai S. On the experimental dengue infection in man. Nippon Igaku 1943; 3325:529 - 31
  • Sabin AB. Research on dengue during World War II. Am J Trop Med Hyg 1952; 1:30 - 50; PMID: 14903434
  • Sabin AB, Schlesinger RW. Production of immunity to dengue with virus modified by propagation in mice. Science 1945; 101:640 - 2; http://dx.doi.org/10.1126/science.101.2634.640; PMID: 17844088
  • McCoy OR, Sabin AB. Dengue. In: Coates JB, Hoff EC, Hoff PM, eds. Preventive Medicine in World War II Communicable Diseases Arthropodborne Diseases Other Than Malaria. Washington, DC: Office of the Surgeon General, 1946:29-62.
  • Hotta S. Experimental studies on dengue. I. Isolation, identification and modification of the virus. J Infect Dis 1952; 90:1 - 9; http://dx.doi.org/10.1093/infdis/90.1.1; PMID: 14888958
  • Yaoi H. A summary of our studies on dengue. Yokohama Med Bull 1958; 9:1 - 20; PMID: 13558217
  • Sun W, Eckels KH, Putnak JR, Lyons AG, Thomas SJ, Vaughn DW, et al. Experimental dengue virus challenge of human subjects previously vaccinated with live attenuated tetravalent dengue vaccines. J Infect Dis 2013; 207:700 - 8; http://dx.doi.org/10.1093/infdis/jis744; PMID: 23225894
  • Gunther VJ, Putnak R, Eckels KH, Mammen MP, Scherer JM, Lyons A, et al. A human challenge model for dengue infection reveals a possible protective role for sustained interferon gamma levels during the acute phase of illness. Vaccine 2011; 29:3895 - 904; http://dx.doi.org/10.1016/j.vaccine.2011.03.038; PMID: 21443963
  • Statler J, Mammen M, Lyons A, Sun W. Sonographic findings of healthy volunteers infected with dengue virus. J Clin Ultrasound 2008; 36:413 - 7; http://dx.doi.org/10.1002/jcu.20476; PMID: 18446859
  • Edelman R, Hombach J. “Guidelines for the clinical evaluation of dengue vaccines in endemic areas”: summary of a World Health Organization Technical Consultation. Vaccine 2008; 26:4113 - 9; http://dx.doi.org/10.1016/j.vaccine.2008.05.058; PMID: 18597906
  • Watanaveeradej V, Simasathien S, Nisalak A, Endy TP, Jarman RG, Innis BL, et al. Safety and immunogenicity of a tetravalent live-attenuated dengue vaccine in flavivirus-naive infants. Am J Trop Med Hyg 2011; 85:341 - 51; http://dx.doi.org/10.4269/ajtmh.2011.10-0501; PMID: 21813857
  • Simasathien S, Thomas SJ, Watanaveeradej V, Nisalak A, Barberousse C, Innis BL, et al. Safety and immunogenicity of a tetravalent live-attenuated dengue vaccine in flavivirus naive children. Am J Trop Med Hyg 2008; 78:426 - 33; PMID: 18337339

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.