Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Volume 19, Number 8—August 2013
Letter

Call to Action for Dengue Vaccine Failure

Cite This Article

To the Editor: Dengue is one of the most widespread infectious diseases globally; transmission now occurs in 128 countries. Although dengue virus (DENV) control strategies have targeted vector control and disease surveillance, the development of an effective vaccine is the holy grail of prevention.

Dengue vaccine development has spanned many decades. A candidate vaccine (Sanofi Pasteur, Swiftwater, PA, USA) containing all 4 DENV serotypes is in advanced clinical testing. However, when given to school children in Thailand, this live-attenuated, tetravalent, dengue–yellow fever 17D chimeric virus vaccine showed major but incomplete efficacy against 3 of the 4 DENV serotypes (DENV 1 [61.2%], DENV-3 [81.3%], and DENV-4 [89.9%]) in the intention-to-treat group but no protection against DENV 2, the most pathogenic of the DENV serotypes (1).

Two observations from the efficacy trial in Thailand provide insights into protective immunity that could greatly improve second-generation vaccines. The first observation was that a single dose of 4 live-attenuated chimeric DENVs given subcutaneously at a single site failed to raise type-specific protective immunity against the 4 DENV serotypes, and 2) doses 2 and 3 of the Sanofi Pasteur vaccine given to children over a 1-year period failed to improve efficacy outcomes. These results were obtained even though 91% of the children had circulating dengue or Japanese encephalitis antibodies before vaccination, neutralizing antibodies developed to all 4 DENVs, and neutralizing antibody titers increased 2–3 fold after 3 doses of vaccine in 80%–90% of vaccinated children.

The inability of a mixture of 4 dengue chimeric viruses to elicit an initial primary neutralizing antibody response in nonhuman primates and susceptible humans was recognized during preclinical testing and explained by the phenomenon of interference (2). Although protective immunity was raised in susceptible rhesus monkeys inoculated with all 4 DENVs at a single site, inoculation of 4 chimeric dengue viruses at 1 or 2 sites did not result in neutralizing antibody responses to all 4 DENV (3). Studies on human primary immune responses to dengue infection have identified critical attachment sites on the virion for neutralizing antibodies (4). Serum samples from children given ≥1 doses of the dengue chimeric vaccine can now be tested for primary neutralizing antibody responses to each of the 4 DENVs. As an alternative, antibody-secreting cells may be isolated and their products identified by using methods as described for dengue-infected children in Nicaragua (5).

Infections with 2 different DENVs can protect against severe disease during subsequent infections (6). It has therefore been assumed that persons with DENV neutralizing antibodies are protected against infection. In clinical testing of the Sanofi Pasteur vaccine, failure of multiple booster doses to show protection was unexpected because the children were already substantially immune from prior exposure to DENV or Japanese encephalitis vaccine. When the tetravalent dengue chimeric vaccine was given to partially dengue–immune children and adults in the Philippines, a broad neutralizing antibody response was observed after administration of only 2 vaccine doses (7).

We believe that the unanticipated results of the dengue vaccine efficacy trial in Thailand call for new methods of assessing dengue immunity. Myeloid cells are major targets of dengue infection in humans. We and others have described the unique biologic responses when dengue virus–antibody complexes are presented to myeloid cells (8). There is evidence that DENV neutralization titers differ when the same antibodies are assayed in epithelial and Fc-receptor–bearing cells (8). Recent work suggests that primary monocytes and macrophages may not respond in exactly the same fashion to infection by DENV immune complexes (8). Few relevant studies exist in the literature, and most focused on DENV-2. Detailed studies on innate immune responses in human myeloid cells with a variety of dengue immune complexes should proceed forthwith.

To our knowledge, only once has an in vitro test correctly predicted which children would be susceptible or have silent infections accompanying a second heterotypic dengue infection (9). This was determined by using serum samples collected before a second dengue infection and testing these serum samples at low dilutions for their ability to protect primary human monocytes from DENV-2 infection or antibody-dependent enhanced infection. During development of the Sanofi Pasteur tetravalent chimeric dengue vaccine, serum samples from vaccinated persons were routinely tested for neutralization of DENV in an epithelial cell line (10). In addition to assaying for antibodies directed at the quaternary site described by de Alwis et al. (4), we suggest that serum samples from vaccinated persons be tested for neutralization of all DENVs in primary human myeloid cells.

Although human Fc-receptor cell lines may be convenient for assaying DENV antibodies, decisions regarding their use should be deferred until they are shown to model primary myeloid cells. Because antibody titers often wane after vaccination, the ability of serum samples from vaccinees to protect against infection of myeloid cells with the 4 DENVs should be studied over many years. Changes to in vitro systems for measuring immune responses after dengue vaccination may provide a better surrogate of protection by realigning antibody measurement systems to our contemporary understanding of the pathogenesis of this complex disease.

Top

Suresh MahalingamComments to Author , Belinda L. Herring, and Scott B. Halstead
Author affiliations: Griffith University, Southport, Queensland, Australia (S. Mahalingam, B.L. Herring); Dengue Vaccine Initiative, Rockville, Maryland, USA (S.B. Halstead)

Top

References

  1. Sabchareon  A, Wallace  D, Sirivichayakul  C, Limkittikul  K, Chanthavanich  P, Suvannadabba  S, Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. Lancet. 2012;380:155967 . DOIPubMedGoogle Scholar
  2. Guy  B, Barban  V, Mantel  N, Aguirre  M, Gulia  S, Pontvianne  J, Evaluation of interferences between dengue vaccine serotypes in a monkey model. Am J Trop Med Hyg. 2009;80:30211 .PubMedGoogle Scholar
  3. Halstead  SB, Casals  J, Shotwell  H, Palumbo  N. Studies on the immunization of monkeys against dengue. I. Protection derived from single and sequential virus infections. Am J Trop Med Hyg. 1973;22:36574 .PubMedGoogle Scholar
  4. de Alwis  R, Smith  SA, Olivarez  NP, Messer  WB, Huynh  JP, Wahala  WM, Identification of human neutralizing antibodies that bind to complex epitopes on dengue virions. Proc Natl Acad Sci U S A. 2012;109:743944. DOIPubMedGoogle Scholar
  5. Zompi  S, Montoya  M, Pohl  MO, Balmaseda  A, Harris  E. Dominant cross-reactive B cell response during secondary acute dengue virus infection in humans. PLos Negl Trop Dis. 2012;6:e1568.
  6. Gibbons  RV, Kalanarooj  S, Jarman  RG, Nisalak  A, Vaughn  DW, Endy  TP, Analysis of repeat hospital admissions for dengue to estimate the frequency of third or fourth dengue infections resulting in admissions and dengue hemorrhagic fever, and serotype sequences. Am J Trop Med Hyg. 2007;77:9103 .PubMedGoogle Scholar
  7. Capeding  RZ, Luna  IA, Bomasang  E, Lupisan  S, Lang  J, Forrat  R, Live-attenuated, tetravalent dengue vaccine in children, adolescents and adults in a dengue endemic country: randomized controlled phase I trial in the Philippines. Vaccine. 2011;29:386372. DOIPubMedGoogle Scholar
  8. Halstead  SB, Mahalingam  S, Marovich  MA, Ubol  S, Mosser  DM. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. Lancet Infect Dis. 2010;10:71222. DOIPubMedGoogle Scholar
  9. Kliks  SC, Nisalak  A, Brandt  WE, Wahl  L, Burke  DS. Antibody-dependent enhancement of dengue virus growth in human monocytes as a risk factor for dengue hemorrhagic fever. Am J Trop Med Hyg. 1989;40:44451 .PubMedGoogle Scholar
  10. Guy  B, Barrere  B, Malinowski  C, Saville  M, Teyssou  R, Lang  J. From research to phase III: preclinical, industrial and clinical development of the Sanofi Pasteur tetravalent dengue vaccine. Vaccine. 2011;29:722941. DOIPubMedGoogle Scholar

Top

Cite This Article

DOI: 10.3201/eid1908.121864

Related Links

Top

Table of Contents – Volume 19, Number 8—August 2013

EID Search Options
presentation_01 Advanced Article Search – Search articles by author and/or keyword.
presentation_01 Articles by Country Search – Search articles by the topic country.
presentation_01 Article Type Search – Search articles by article type and issue.

Top

Comments

Please use the form below to submit correspondence to the authors or contact them at the following address:

Suresh Mahalingam, Institute for Glycomics, Griffith University, Parksland Dr, Gold Coast Campus, Southport, Queensland 4222, Australia

Send To

10000 character(s) remaining.

Top

Page created: July 19, 2013
Page updated: July 19, 2013
Page reviewed: July 19, 2013
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
file_external