Skip to main content
The Journal of Infectious Diseases logoLink to The Journal of Infectious Diseases
editorial
. 2011 Feb 1;203(3):299–303. doi: 10.1093/infdis/jiq060

The Necessity and Quandaries of Dengue Vaccine Development

Stephen J Thomas 1,
PMCID: PMC3071120  PMID: 21208919

(See the article by Durbin et al, on pages 327–334)

Dengue is an emerging and reemerging arboviral disease of great global public health importance. Increased transmission and disease outbreaks are being driven by population growth, urbanization, international travel, and unchecked vector populations [1]. Southeast Asia, Central and South America, and parts of the Caribbean experience endemic and hyperendemic dengue virus (DENV) transmission while indigenous transmission is being increasingly recognized in areas of Africa, the Middle East and South Asia [25]. Reports indicate that southern US border-states and Hawaii can support episodic DENV transmission [612]. Dengue poses a risk to traveler and military populations, especially those originating from non-dengue endemic regions [1318].

Millions of DENV infections, hundreds of thousands of hospitalizations, and tens of thousands of deaths related to dengue occur annually [19]. There is no specific, licensed anti-DENV therapeutic or preventative vaccine. The financial, social and individual cost of dengue is significant, underestimated, and underappreciated [2025]. The strategic administration of a safe and efficacious dengue vaccine, in coordination with efforts to educate about personal protective measures and sustained vector control, is the best hope to reduce the global dengue burden.

There are numerous dengue vaccine candidates in clinical development. Early efforts to develop a dengue vaccine date back more than 70 years, with attempts to prevent virus transmission using infectious human plasma treated with ox bile or virus grown in live mosquitoes and inactivated with formalin [26]. Schelsinger and Sabin undertook the first attempts to immunize using mouse-passaged live-attenuated DENV-1 and -2 viruses [2729]. Halstead and colleagues discovered DENVs were attenuated following passage in primary dog kidney (PDK) cell culture [30]. Mahidol University and Sanofi Pasteur attempted to codevelop live attenuated virus dengue vaccine candidates using PDK cell passage; the Walter Reed Army Institute of Research and GlaxoSmithKline Biologicals also used PDK passage to attenuate vaccine virus strain candidates [3140]. The US National Institutes of Allergy and Infectious Diseases attenuate DENV strains by targeted mutagenesis; the resulting attenuated DENV strains may constitute stand-alone vaccine candidates or serve as chimeric backbones [4145]. The US Naval Medical Research Center has completed a phase 1 trial testing a DENV-1 pre-Membrane/Envelope DNA vaccine; explorations of different vector and/or adjuvant combinations continue [4650]. Hawaii Biotech/Merck & Company is completing a phase 1 trial testing a DENV-1 recombinant Envelope protein candidate [5153]. Sanofi Pasteur is in advanced clinical development (phase 3) of a chimeric-Yellow fever-dengue (CYD) vaccine candidate using a construct created at the St. Louis University Health Sciences Center and Acambis Inc. [5459]. The CYD candidate is the first candidate to enter clinical endpoint trials.

Efforts to develop a dengue vaccine have been plagued by numerous challenges and quandaries, some established and others hypothetical. The most obvious quandary for the dengue vaccinologist is the existence of 4 DENV types, each capable of causing severe dengue and death. The global epidemiology of dengue and cocirculation of multiple DENV types within tight geographic areas mandates the need for a vaccine capable of protecting against disease caused by any DENV type, a tetravalent vaccine (ie, containing DENV-1-4 antigens) [2, 60]. The implication of this requirement extends well beyond the need to produce 4 DENV monovalent vaccines and then successfully combine into a single tetravalent formulation. Human and non-human primate studies have demonstrated the existence of DENV type-specific immune dominance/interference when administering tetravalent vaccine candidates, with the result being an imbalanced immune response, a very undesirable outcome (discussed below) [61, 62]. Whether this is a phenomenon and challenge only for live virus replicating vaccines or all tetravalent dengue vaccines is unknown.

The absence of a validated animal model of dengue disease is also a challenge to the vaccine development effort. Research in this area continues but at this time vaccine developers are without a reproducible and relevant disease model to initially test vaccine candidates, down-select promising formulations, and understand, early on, the potential for clinical benefit [6367]. As a result, numerous, small-scale phase 1 and 2 human trials are required to advance candidates.

In addition to the absence of an animal model, there is no validated human challenge model. Timelines for developing and down-selecting malaria vaccine candidates have benefitted greatly from the Plasmodium falciparum human challenge model developed at the WRAIR [68]. Experimental human infection with DENV has been reproduced in hundreds over the last century without untoward effects [2628, 6973]. During the past decade the WRAIR has attempted to validate minimally attenuated DENV-1–4 vaccine candidates as human challenge strains with limited success [74]. The requirement for cGMP manufacture and complex regulatory and human subjects’ protection requirements make this pursuit very resource intensive. It is unclear how a dengue human challenge model would need to perform (ie, reproduce dengue fever versus reproduce viremia with or without symptoms) to support dengue vaccine development plans.

The incomplete understanding of what “immune profile” will lead to a protective or pathogenic response following a DENV infection poses another challenge to developers. Although anti-DENV neutralizing antibodies are likely required for protection from dengue disease, it is well established certain antibody characteristics (ie, non-neutralizing, cross-reactive, low affinity) may contribute to a poor clinical outcome [7580]. Additionally, cellular immunity plays a role in both protective and pathogenic outcomes following exposure [8185]. Dengue vaccine candidates would, ideally, induce immune responses corresponding to protective, rather than pathogenic, profiles. The spectrum of immune profiles induced by dengue vaccine candidates using different approaches (eg, live virus, chimeras, DNA) is unclear, but vaccine developers are devoting resources to broadly characterize responses [8692].

There is no established dengue immune correlate of protection. An immune correlate would support (1) understanding how vaccine immunogenicity relates to protection from disease; (2) generalizing efficacy across different populations; (3) facilitating bridging between clinical studies; and (4) defining the relevant parameter to establish vaccine potency tests [93]. Without a validated human challenge model, attempts to define an immune correlate will need to be made in the context of clinical endpoint trials.

The dengue vaccine field is also challenged by the biologic assays currently available to measure immunogenicity. The measurement of neutralizing antibody is the most relevant endpoint to the vaccine development effort from a scientific and regulatory perspective; neutralizing antibody is believed to be protective and is consistently measurable. Variations on the plaque reduction neutralization test (PRNT) or microneutralization assay platforms are currently used to measure neutralizing antibody [94, 95]. Unfortunately, assay results can be variable and, in the face of multiple antigen exposure (ie, secondary infection or vaccination with tetravalent dengue vaccines), difficult to interpret whether homotypic, high-quality antibody (ie, neutralizing and protective) or cross-reactive antibody is being measured [96]. Furthermore, each developer utilizes methods and reagents specific for their vaccine candidate, making inter-developer immunogenicity comparisons nearly impossible [97]. The Pediatric Dengue Vaccine Initiative and World Health Organization have attempted to facilitate standardization and harmonization of the PRNT across laboratories [95]. Dengue vaccine efficacy trials and associations between neutralizing antibody measurements and various clinical outcomes will improve our understanding of immunogenicity endpoints.

A major theoretical concern is that poorly immunizing vaccines (ie, low antibody titer, no induction of T or B cell memory), imbalanced responses (ie, variable antibody responses to each DENV type), or waning immunity (ie, decline in antibody titer over time) may increase vaccine recipient risk of an immunopathologic response (ie, enhanced disease) following subsequent natural infection or re-immunization [60]. Limited, long-term studies of dengue vaccine recipients residing in dengue-endemic areas and being exposed to natural infections have not revealed an increased risk of severe disease [98]. In this issue of the Journal, Durbin and colleagues [99] describe the results of experiments exposing recipients of attenuated monovalent dengue vaccines to heterotypic monovalent vaccines 0.6–7.4 years later. There were no overt safety signals observed compared with control (dengue vaccination in naïve volunteers), and neutralizing antibody profiles in the heterotypic group were broad, qualitatively mimicking what is seen in natural secondary dengue virus infections. There are limitations in equating these experiments with natural secondary DENV infections (ie, higher risk for severe disease) or extrapolating results to potential dengue vaccine recipients who are primed with a natural DENV or another flavivirus (ie, Yellow fever virus or Japanese encephalitis virus), but the investigators provide an important step in assessing the safety of vaccinating DENV immune subjects. Early dengue vaccine efficacy trials will require long-term subject follow-up or phase 4 studies to better define the risks of immunizing populations in dengue endemic areas and areas where other flaviviruses circulate [60].

The global burden of dengue is significant and the world needs a dengue vaccine. The recent infusion of financial resources into dengue research greatly expanded what was once a narrow field of dengue scientists and funding entities. There are many dengue vaccine development initiatives underway, with numerous candidates in preclinical and clinical development. Data from the first dengue vaccine efficacy trial are greatly anticipated. Challenges to the vaccine development effort exist and there is much to learn. The worsening financial and societal burden of dengue calls for increased funding to facilitate the study, and improved understanding of dengue epidemiology, immunology, and the development and advancement of vaccine candidates with the potential to provide clinical benefit.

Funding

No funding was received in support of this editorial.

Acknowledgments

The author is grateful to Dr Timothy P. Endy and Dr In-Kyu Yoon for sharing their perspectives on the global dengue problem and providing helpful guidance for this editorial.

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

References

  • 1.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. doi: 10.1016/s0966-842x(01)02288-0. [DOI] [PubMed] [Google Scholar]
  • 2.Thomas SJ, Strickman D, Vaughn DW. Dengue epidemiology: virus epidemiology, ecology, and emergence. Adv Virus Res. 2003;61:235–89. doi: 10.1016/s0065-3527(03)61006-7. [DOI] [PubMed] [Google Scholar]
  • 3.Ayyub M, Khazindar AM, Lubbad EH, Barlas S, Alfi AY, Al-Ukayli S. Characteristics of dengue fever in a large public hospital, Jeddah, Saudi Arabia. J Ayub Med Coll Abbottabad. 2006;18:9–13. [PubMed] [Google Scholar]
  • 4.Ahmed S, Arif F, Yahya Y, et al. Dengue fever outbreak in Karachi 2006–a study of profile and outcome of children under 15 years of age. J Pak Med Assoc. 2008;58:4–8. [PubMed] [Google Scholar]
  • 5.Dorji T, Yoon IK, Holmes EC, et al. Diversity origin of dengue virus serotypes 1, 2, and 3, Bhutan. Emerg Infect Dis. 2009;15:1630–2. doi: 10.3201/eid1510.090123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Effler PV, Pang L, Kitsutani P, et al. Dengue fever, Hawaii, 2001–2002. Emerg Infect Dis. 2005;11:742–9. doi: 10.3201/eid1105.041063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ramos MM, Mohammed H, Zielinski-Gutierrez E, et al. Epidemic dengue dengue hemorrhagic fever at the Texas-Mexico border: results of a household-based seroepidemiologic survey, December 2005. Am J Trop Med Hyg. 2008;78:364–9. [PubMed] [Google Scholar]
  • 8.Dengue hemorrhagic fever–U.S.-Mexico border, 2005. Morb Mortal Wkly Rep. 2007;56:785–9. [PubMed] [Google Scholar]
  • 9.From the Centers for Disease Control Prevention. Dengue fever at the US-Mexico border, 1995–1996. JAMA. 1996;276:1464–65. [PubMed] [Google Scholar]
  • 10.Rigau-Perez JG, Gubler DJ, Vorndam AV, Clark GG. Dengue surveillance–United states, 1986–1992. MMWR CDC Surveill Summ. 1994;43:7–19. [PubMed] [Google Scholar]
  • 11.Morens DM, Fauci AS. Dengue and hemorrhagic fever: a potential threat to public health in the United States. JAMA. 2008;299:214–216. doi: 10.1001/jama.2007.31-a. [DOI] [PubMed] [Google Scholar]
  • 12.Locally acquired Dengue–Key West, Florida, 2009-2010. Morb Mortal Wkly Rep. 2010;59:577–81. [PubMed] [Google Scholar]
  • 13.Laferl H, Szell M, Bischof E, Wenisch C. Imported dengue fever in Austria 1990-2005. Travel Med Infect Dis. 2006;4:319–23. doi: 10.1016/j.tmaid.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 14.Wilder-Smith A, Schwartz E. Dengue in travelers. N Engl J Med. 2005;353:924–32. doi: 10.1056/NEJMra041927. [DOI] [PubMed] [Google Scholar]
  • 15.Travel-associated dengue infections–United States, 2001–2004. Morb Mortal Wkly Rep. 2005;54:556–58. [PubMed] [Google Scholar]
  • 16.Sung V, O'Brien DP, Matchett E, Brown GV, Torresi J. Dengue Fever in travelers returning from southeast Asia. J Travel Med. 2003;10:208–13. doi: 10.2310/7060.2003.40555. [DOI] [PubMed] [Google Scholar]
  • 17.Trofa AF, DeFraites RF, Smoak BL, et al. Dengue fever in US military personnel in Haiti. JAMA. 1997;277:1546–48. [PubMed] [Google Scholar]
  • 18.From the Centers for Disease Control Prevention. Dengue fever among U.S. military personnel–Haiti, September-November, 1994. JAMA. 1995;273:14–15. [PubMed] [Google Scholar]
  • 19.Dengue and dengue haemorrhagic fever. Available at: http://www.who.int/mediacentre/factsheets/fs117/en/. Accessed 10 October 2010 2010. [Google Scholar]
  • 20.Luz PM, Grinsztejn B, Galvani AP. Disability adjusted life years lost to dengue in Brazil. Trop Med Int Health. 2009;14:237–46. doi: 10.1111/j.1365-3156.2008.02203.x. [DOI] [PubMed] [Google Scholar]
  • 21.Huy R, Wichmann O, Beatty M, et al. Cost of dengue and other febrile illnesses to households in rural Cambodia: a prospective community-based case-control study. BMC Public Health. 2009;9:155. doi: 10.1186/1471-2458-9-155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Garg P, Nagpal J, Khairnar P, Seneviratne SL. Economic burden of dengue infections in India. Trans R Soc Trop Med Hyg. 2008;102:570–77. doi: 10.1016/j.trstmh.2008.02.015. [DOI] [PubMed] [Google Scholar]
  • 23.Torres JR, Castro J. The health and economic impact of dengue in Latin America. Cad Saude Publica. 2007;23:S23–S31. doi: 10.1590/s0102-311x2007001300004. [DOI] [PubMed] [Google Scholar]
  • 24.Anderson KB, Chunsuttiwat S, Nisalak A, et al. Burden of symptomatic dengue infection in children at primary school in Thailand: a prospective study. Lancet. 2007;369:1452–59. doi: 10.1016/S0140-6736(07)60671-0. [DOI] [PubMed] [Google Scholar]
  • 25.Clark DV, Mammen MP, Jr, Nisalak A, Puthimethee V, Endy TP. Economic impact of dengue fever/dengue hemorrhagic fever in Thailand at the family and population levels. Am J Trop Med Hyg. 2005;72:786–91. [PubMed] [Google Scholar]
  • 26.Simmons JS, St John JH, Reynolds FHK. Experimental studies of dengue. Philippine J Sci. 1931;44:1–252. [Google Scholar]
  • 27.Schlesinger RW, Gordon I, Frankel JW, Winter JW, Patterson PR, Dorrance WR. Clinical and serologic response of man to immunization with attnuated dengue and yellow fever viruses. JImmunol. 1956;77:352–64. [PubMed] [Google Scholar]
  • 28.Sabin AB, Schlesinger RW. Production of immunity to dengue with virus modified by propagation in mice. Science (New York, NY) 1945;101:640–42. doi: 10.1126/science.101.2634.640. [DOI] [PubMed] [Google Scholar]
  • 29.Wisseman CL, Jr, Sweet BH, Rosenzweig EC, Eylar OR. Attenuated living type 1 dengue vaccines. Am J Trop Med Hyg. 1963;12:620–23. [Google Scholar]
  • 30.Halstead SB, Diwan AR, Marchette NJ, Palumbo NE, Srisukonth L. Selection of attenuated dengue 4 viruses by serial passage in primary kidney cells. I. Attributes of uncloned virus at different passage levels. Am J Trop Med Hyg. 1984;33:654–65. doi: 10.4269/ajtmh.1984.33.654. [DOI] [PubMed] [Google Scholar]
  • 31.Bhamarapravati N, Yoksan S, Chayaniyayothin T, Angsubphakorn S, Bunyaratvej A. Immunization with a live attenuated dengue-2-virus candidate vaccine (16681-PDK 53): clinical, immunological and biological responses in adult volunteers. Bull World Health Organ. 1987;65:189–95. [PMC free article] [PubMed] [Google Scholar]
  • 32.Bhamarapravati N, Sutee Y. Live attenuated tetravalent dengue vaccine. Vaccine. 2000;18:44–7. doi: 10.1016/s0264-410x(00)00040-2. [DOI] [PubMed] [Google Scholar]
  • 33.Sabchareon A, Lang J, Chanthavanich P, et al. Safety and immunogenicity of a three dose regimen of two tetravalent live-attenuated dengue vaccines in five- to twelve-year-old Thai children. Pediatr Infect Dis J. 2004;23:99–109. doi: 10.1097/01.inf.0000109289.55856.27. [DOI] [PubMed] [Google Scholar]
  • 34.Sabchareon A, Lang J, Chanthavanich P, et al. Safety and immunogenicity of tetravalent live-attenuated dengue vaccines in Thai adult volunteers: role of serotype concentration, ratio, and multiple doses. Am J Trop Med Hyg. 2002;66:264–72. doi: 10.4269/ajtmh.2002.66.264. [DOI] [PubMed] [Google Scholar]
  • 35.Sun W, Cunningham D, Wasserman SS, et al. Phase 2 clinical trial of three formulations of tetravalent live-attenuated dengue vaccine in flavivirus-naive adults. Hum Vaccin. 2009;5:33–40. doi: 10.4161/hv.5.1.6348. [DOI] [PubMed] [Google Scholar]
  • 36.Sun W, Edelman R, Kanesa-Thasan N, et al. Vaccination of human volunteers with monovalent tetravalent live-attenuated dengue vaccine candidates. Am J Trop Med Hyg. 2003;69:24–31. doi: 10.4269/ajtmh.2003.69.6_suppl.0690024. [DOI] [PubMed] [Google Scholar]
  • 37.Kanesa-Thasan N, Edelman R, Tacket CO, et al. Phase 1 studies of Walter Reed Army Institute of Research candidate attenuated dengue vaccines: selection of safe and immunogenic monovalent vaccines. Am J Trop Med Hyg. 2003;69:17–23. doi: 10.4269/ajtmh.2003.69.17. [DOI] [PubMed] [Google Scholar]
  • 38.Innis BL, Eckels KH. Progress in development of a live-attenuated, tetravalent dengue virus vaccine by the United States Army Medical Research and Materiel Command. Am J Trop Med Hyg. 2003;69:1–4. doi: 10.4269/ajtmh.2003.69.6_suppl.0690001. [DOI] [PubMed] [Google Scholar]
  • 39.Eckels KH, Dubois DR, Putnak R, et al. Modification of dengue virus strains by passage in primary dog kidney cells: preparation of candidate vaccines and immunization of monkeys. Am J Trop Med Hyg. 2003;69:12–6. doi: 10.4269/ajtmh.2003.69.12. [DOI] [PubMed] [Google Scholar]
  • 40.Simasathien S, Thomas SJ, Watanaveeradej V, 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. [PubMed] [Google Scholar]
  • 41.Durbin AP, Karron RA, Sun W, et al. Attenuation and immunogenicity in humans of a live dengue virus type-4 vaccine candidate with a 30 nucleotide deletion in its 3′-untranslated region. Am J Trop Med Hyg. 2001;65:405–13. doi: 10.4269/ajtmh.2001.65.405. [DOI] [PubMed] [Google Scholar]
  • 42.Blaney JE, Jr, Durbin AP, Murphy BR, Whitehead SS. Targeted mutagenesis as a rational approach to dengue virus vaccine development. Curr Top Microbiol Immunol. 2010;338:145–58. doi: 10.1007/978-3-642-02215-9_11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wright PF, Durbin AP, Whitehead SS, et al. Phase 1 trial of the dengue virus type 4 vaccine candidate rDEN4{Delta}30-4995 in healthy adult volunteers. Am J Trop Med Hyg. 2009;81:834–41. doi: 10.4269/ajtmh.2009.09-0131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Durbin AP, McArthur JH, Marron JA, et al. rDEN2/4Delta30(ME), a live attenuated chimeric dengue serotype 2 vaccine is safe highly immunogenic in healthy dengue-naive adults. Hum Vaccin. 2006;2:255–60. doi: 10.4161/hv.2.6.3494. [DOI] [PubMed] [Google Scholar]
  • 45.Blaney JE, Jr, Durbin AP, Murphy BR, Whitehead SS. Development of a live attenuated dengue virus vaccine using reverse genetics. Viral Immunol. 2006;19:10–32. doi: 10.1089/vim.2006.19.10. [DOI] [PubMed] [Google Scholar]
  • 46.Safety study of a dengue virus DNA vaccine. Available at: http://clinicaltrials.gov/ct2/show/NCT00290147?term=dengue+and+dna&rank=1. Accessed October 12, 2010 2010. [Google Scholar]
  • 47.Raviprakash K, Apt D, Brinkman A, et al. A chimeric tetravalent dengue DNA vaccine elicits neutralizing antibody to all four virus serotypes in rhesus macaques. Virology. 2006;353:166–73. doi: 10.1016/j.virol.2006.05.005. [DOI] [PubMed] [Google Scholar]
  • 48.Raviprakash K, Kochel TJ, Ewing D, et al. Immunogenicity of dengue virus type 1 DNA vaccines expressing truncated and full length envelope protein. Vaccine. 2000;18:2426–34. doi: 10.1016/s0264-410x(99)00570-8. [DOI] [PubMed] [Google Scholar]
  • 49.Raviprakash K, Porter KR, Kochel TJ, et al. Dengue virus type 1 DNA vaccine induces protective immune responses in rhesus macaques. J Gen Virol. 2000;81:1659–67. doi: 10.1099/0022-1317-81-7-1659. [DOI] [PubMed] [Google Scholar]
  • 50.Raviprakash K, Wang D, Ewing D, et al. A tetravalent dengue vaccine based on a complex adenovirus vector provides significant protection in rhesus monkeys against all four serotypes of dengue virus. J Virol. 2008 Jul;82(14):6927–34. doi: 10.1128/JVI.02724-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Clements DE, Coller BA, Lieberman MM, et al. Development of a recombinant tetravalent dengue virus vaccine: immunogenicity and efficacy studies in mice and monkeys. Vaccine. 2010;28:2705–15. doi: 10.1016/j.vaccine.2010.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Putnak R, Coller BA, Voss G, et al. An evaluation of dengue type-2 inactivated, recombinant subunit, and live-attenuated vaccine candidates in the rhesus macaque model. Vaccine. 2005;23:4442–52. doi: 10.1016/j.vaccine.2005.03.042. [DOI] [PubMed] [Google Scholar]
  • 53.Study of HBV-001 D1 in healthy adults. Available at: http://clinicaltrials.gov/ct2/show/NCT00936429?term=hawaii+biotech&rank=2. Accessed 12 October 2010. [Google Scholar]
  • 54.Jones T. Technology evaluationVax-DEN, Acambis/Aventis. Curr Opin Mol Ther. 2004;6:443–50. [PubMed] [Google Scholar]
  • 55.Guy B, Guirakhoo F, Barban V, Higgs S, Monath TP, Lang J. Preclinical and clinical development of YFV 17D-based chimeric vaccines against dengue, West Nile and Japanese encephalitis viruses. Vaccine. 2010;28:632–49. doi: 10.1016/j.vaccine.2009.09.098. [DOI] [PubMed] [Google Scholar]
  • 56.Guirakhoo F, Kitchener S, Morrison D, et al. Live attenuated chimeric yellow fever dengue type 2 (ChimeriVax-DEN2) vaccine: phase I clinical trial for safety and immunogenicity: effect of yellow fever pre-immunity in induction of cross neutralizing antibody responses to all 4 dengue serotypes. Hum Vaccin. 2006;2:60–7. doi: 10.4161/hv.2.2.2555. [DOI] [PubMed] [Google Scholar]
  • 57.Guy B, Saville M, Lang J. Development of Sanofi Pasteur tetravalent dengue vaccine. Hum Vaccin. 2010;6 doi: 10.4161.hv.6.9.12739. [DOI] [PubMed] [Google Scholar]
  • 58.Lang J. Recent progress on sanofi pasteur's dengue vaccine candidate. J Clin Virol. 2009;46:S20–S24. doi: 10.1016/S1386-6532(09)70291-4. [DOI] [PubMed] [Google Scholar]
  • 59.Morrison D, Legg TJ, Billings CW, Forrat R, Yoksan S, Lang J. A novel tetravalent dengue vaccine is well tolerated and immunogenic against all 4 serotypes in flavivirus-naive adults. J Infect Dis. 2010;201:370–7. doi: 10.1086/649916. [DOI] [PubMed] [Google Scholar]
  • 60.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;26:4113–19. doi: 10.1016/j.vaccine.2008.05.058. [DOI] [PubMed] [Google Scholar]
  • 61.Guy B, Barban V, Mantel N, et al. Evaluation of interferences between dengue vaccine serotypes in a monkey model. Am J Trop Med Hyg. 2009;80:302–11. [PubMed] [Google Scholar]
  • 62.Edelman R, Wasserman SS, Bodison SA, et al. Phase I trial of 16 formulations of a tetravalent live-attenuated dengue vaccine. Am J Trop Med Hyg. 2003;69:48–60. doi: 10.4269/ajtmh.2003.69.48. [DOI] [PubMed] [Google Scholar]
  • 63.Shresta S, Sharar KL, Prigozhin DM, Beatty PR, Harris E. Murine model for dengue virus-induced lethal disease with increased vascular permeability. J Virol. 2006 Oct;80(20):10208–17. doi: 10.1128/JVI.00062-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Williams KL, Zompi S, Beatty PR, Harris E. A mouse model for studying dengue virus pathogenesis and immune response. Ann N Y Acad Sci. 2009;1171:E12–E23. doi: 10.1111/j.1749-6632.2009.05057.x. [DOI] [PubMed] [Google Scholar]
  • 65.Cassetti MC, Durbin A, Harris E, Rico-Hesse R, et al. Report of an NIAID workshop on dengue animal models. Vaccine. 2010;28:4229–34. doi: 10.1016/j.vaccine.2010.04.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Onlamoon N, Noisakran S, Hsiao HM, et al. Dengue virus-induced hemorrhage in a nonhuman primate model. Blood. 2010;115:1823–34. doi: 10.1182/blood-2009-09-242990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tan GK, Ng JK, Trasti SL, Schul W, Yip G, Alonso S. A non mouse-adapted dengue virus strain as a new model of severe dengue infection in AG129 mice. PLoS Negl Trop Dis. 2010;4:e672. doi: 10.1371/journal.pntd.0000672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Chulay JD, Schneider I, Cosgriff TM, et al. Malaria transmitted to humans by mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg. 1986;35:66–8. doi: 10.4269/ajtmh.1986.35.66. [DOI] [PubMed] [Google Scholar]
  • 69.Ashburn PM, Craig CF. Experimental investigations regarding the etiology of dengue fever. J Infect Dis. 1907;4:440–75. doi: 10.1086/383418. [DOI] [PubMed] [Google Scholar]
  • 70.Siler JF, Hall MW, Hitchens AP. Dengue: its history, epidemilogy, mechanism of transmission, etiology, clinical manifestations, immunity, and prevention. Philippine J Sci. 1926;29:1–304. [Google Scholar]
  • 71.Hotta S. Experimental studies on dengue. I. Isolation, identification and modification of the virus. J Infect Dis. 1952;90:1–9. doi: 10.1093/infdis/90.1.1. [DOI] [PubMed] [Google Scholar]
  • 72.Innis BL, Eckels KH, Kraiselburd E, et al. Virulence of a live dengue virus vaccine candidate: a possible new marker of dengue virus attenuation. J Infect Dis. 1988;158:876–80. doi: 10.1093/infdis/158.4.876. [DOI] [PubMed] [Google Scholar]
  • 73.McKee KT, Jr, Bancroft WH, Eckels KH, Redfield RR, Summers PL, Russell PK. Lack of attenuation of a candidate dengue 1 vaccine (45AZ5) in human volunteers. Am J Trop Med Hyg. 1987;36:435–42. doi: 10.4269/ajtmh.1987.36.435. [DOI] [PubMed] [Google Scholar]
  • 74.Statler J, Mammen M, Lyons A, Sun W. Sonographic findings of healthy volunteers infected with dengue virus. J Clin Ultrasound. 2008;36:413–17. doi: 10.1002/jcu.20476. [DOI] [PubMed] [Google Scholar]
  • 75.Halstead SB. In vivo enhancement of dengue virus infection in Rhesus monkeys by passively transferred antibody. J Infect Dis. 1979;140:527–33. doi: 10.1093/infdis/140.4.527. [DOI] [PubMed] [Google Scholar]
  • 76.Halstead SB, Porterfield JS, O'Rourke EJ. Enhancement of dengue virus infection in monocytes by flavivirus antisera. Am J Trop Med Hyg. 1980;29:638–42. doi: 10.4269/ajtmh.1980.29.638. [DOI] [PubMed] [Google Scholar]
  • 77.Kurane I, Mady BJ, Ennis FA. Antibody-dependent enhancement of dengue virus infection. Rev Med Virol. 1991;1:211–21. [Google Scholar]
  • 78.Burke DS, Kliks S. Antibody-dependent enhancement in dengue virus infections. J Infect Dis. 2006;193:601–3. doi: 10.1086/499282. author reply 603–4. [DOI] [PubMed] [Google Scholar]
  • 79.Libraty DH, Acosta LP, Tallo V, et al. A prospective nested case-control study of Dengue in infants: rethinking and refining the antibody-dependent enhancement dengue hemorrhagic fever model. PLoS Med. 2009;6:e1000171. doi: 10.1371/journal.pmed.1000171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dejnirattisai W, Jumnainsong A, Onsirisakul N, et al. Cross-reacting antibodies enhance dengue virus infection in humans. Science (New York, NY) 2010;328:745–48. doi: 10.1126/science.1185181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Simmons CP, Dong T, Chau NV, et al. Early T-cell responses to dengue virus epitopes in Vietnamese adults with secondary dengue virus infections. J virology. 2005;79:5665–75. doi: 10.1128/JVI.79.9.5665-5675.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rothman AL. Cellular immunology of sequential dengue virus infection and its role in disease pathogenesis. Curr Top Microbiol Immunol. 2010;338:83–98. doi: 10.1007/978-3-642-02215-9_7. [DOI] [PubMed] [Google Scholar]
  • 83.Friberg H, Burns L, Woda M, et al. Memory CD8(+) T cells from naturally acquired primary dengue virus infection are highly cross-reactive. Immunol Cell Biol. 2010 doi: 10.1038/icb.2010.61. Apr 27. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mathew A, Rothman AL. Understanding the contribution of cellular immunity to dengue disease pathogenesis. Immunol Rev. 2008;225:300–13. doi: 10.1111/j.1600-065X.2008.00678.x. [DOI] [PubMed] [Google Scholar]
  • 85.Green S, Rothman A. Immunopathological mechanisms in dengue and dengue hemorrhagic fever. Curr Opin Infect Dis. 2006;19:429–36. doi: 10.1097/01.qco.0000244047.31135.fa. [DOI] [PubMed] [Google Scholar]
  • 86.Sanchez V, Gimenez S, Tomlinson B, et al. Innate and adaptive cellular immunity in flavivirus-naive human recipients of a live-attenuated dengue serotype 3 vaccine produced in Vero cells (VDV3) Vaccine. 2006;24:4914–26. doi: 10.1016/j.vaccine.2006.03.066. [DOI] [PubMed] [Google Scholar]
  • 87.Rabablert J, Dharakul T, Yoksan S, Bhamarapravati N. Dengue virus specific T cell responses to live attenuated monovalent dengue-2 and tetravalent dengue vaccines. Asian Pac J Allergy Immunol/launched by Allergy Immunol Soc Thailand. 2000;18:227–35. [PubMed] [Google Scholar]
  • 88.Rabablert J, Yoksan S. Attenuated D2 16681-PDK53 vaccine: defining humoral and cell-mediated immunity. Curr Pharm Des. 2009;15:1203–11. doi: 10.2174/138161209787846865. [DOI] [PubMed] [Google Scholar]
  • 89.Green S, Kurane I, Edelman R, et al. Dengue virus-specific human CD4+ T-lymphocyte responses in a recipient of an experimental live-attenuated dengue virus type 1 vaccine: bulk culture proliferation, clonal analysis, and precursor frequency determination. J Virol. 1993;67:5962–7. doi: 10.1128/jvi.67.10.5962-5967.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Guy B. Immunogenicity of sanofi pasteur tetravalent dengue vaccine. J Clin Virol. 2009;46:S16–S19. doi: 10.1016/S1386-6532(09)70290-2. [DOI] [PubMed] [Google Scholar]
  • 91.Guy B, Nougarede N, Begue S, et al. Cell-mediated immunity induced by chimeric tetravalent dengue vaccine in naive or flavivirus-primed subjects. Vaccine. 2008;26:5712–21. doi: 10.1016/j.vaccine.2008.08.019. [DOI] [PubMed] [Google Scholar]
  • 92.Rothman AL, Kanesa-thasan N, West K, Janus J, Saluzzo JF, Ennis FA. Induction of T lymphocyte responses to dengue virus by a candidate tetravalent live attenuated dengue virus vaccine. Vaccine. 2001;19:4694–99. doi: 10.1016/s0264-410x(01)00236-5. [DOI] [PubMed] [Google Scholar]
  • 93.Hombach J, Cardosa MJ, Sabchareon A, Vaughn DW, Barrett AD. Scientific consultation on immunological correlates of protection induced by dengue vaccines report from a meeting held at the World Health Organization 17-18 November 2005. Vaccine. 2007;25:4130–39. doi: 10.1016/j.vaccine.2007.02.079. [DOI] [PubMed] [Google Scholar]
  • 94.Vorndam V, Beltran M. Enzyme-linked immunosorbent assay-format microneutralization test for dengue viruses. Am J Trop Med Hyg. 2002;66:208–12. doi: 10.4269/ajtmh.2002.66.208. [DOI] [PubMed] [Google Scholar]
  • 95.Roehrig JT, Hombach J, Barrett AD. Guidelines for plaque-reduction neutralization testing of human antibodies to dengue viruses. Viral Immunol. 2008;21:123–32. doi: 10.1089/vim.2008.0007. [DOI] [PubMed] [Google Scholar]
  • 96.Putnak JR, de la Barrera R, Burgess T, et al. Comparative evaluation of three assays for measurement of dengue virus neutralizing antibodies. Am J Trop Med Hyg. 2008;79:115–22. [PubMed] [Google Scholar]
  • 97.Thomas SJ, Nisalak A, Anderson KB, et al. Dengue plaque reduction neutralization test (PRNT) in primary secondary dengue virus infections: how alterations in assay conditions impact performance. Am J Trop Med Hyg. 2009;81:825–33. doi: 10.4269/ajtmh.2009.08-0625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Chanthavanich P, Luxemburger C, Sirivichayakul C, et al. Short report: immune response occurrence of dengue infection in thai children three to eight years after vaccination with live attenuated tetravalent dengue vaccine. Am J Trop Med Hyg. 2006;75:26–8. doi: 10.4269/ajtmh.2006.75.1.0750026. [DOI] [PubMed] [Google Scholar]
  • 99. Durbin et al. J Infect Dis 2011; 203:327–334 (in this issue) [Google Scholar]

Articles from The Journal of Infectious Diseases are provided here courtesy of Oxford University Press

RESOURCES