Skip to main content
PLOS Neglected Tropical Diseases logoLink to PLOS Neglected Tropical Diseases
. 2025 Sep 22;19(9):e0013550. doi: 10.1371/journal.pntd.0013550

Antibody responses to Japanese encephalitis virus and dengue virus serotype 2 in children from an orthoflavivirus endemic region after IMOJEV vaccination

Fatima Ericka S Vista 1,2,3,*, Leslie Michelle M Dalmacio 1, Pauline R Solis 4, Cecilia Nelia C Maramba-Lazarte 5,6, Diane M Lang 2,3, Alan L Rothman 2,3, Sheriah Laine M de Paz-Silava 7
Editor: Joshua Anzinger8
PMCID: PMC12463327  PMID: 40982537

Abstract

Background

Japanese encephalitis virus (JEV) is a mosquito-borne pathogen that causes severe neurologic disease. Its endemicity in Asia has prompted its inclusion in nationwide immunization programs. However, the Philippines, which is also endemic for related viruses like dengue (DENV), has not yet adopted this practice. Vaccine hesitancy is a major challenge, exacerbated by concerns over cross-reactive antibodies that may enhance viral infection. This study aimed to determine whether IMOJEV vaccination would induce cross-neutralizing or enhancing antibodies against DENV.

Methodology/Principal findings

Pre- and one-month post-vaccination samples from IMOJEV-vaccinated Filipino children (9–24 months old) were analyzed. A reporter virus particle (RVP)-based neutralization assay against JEV showed neutralization in 28/29 subjects post-vaccination. Presence of DENV2-reactive antibodies was measured via DENV2 VLP ELISA, which revealed increased DENV2 binding reactivity post-vaccination. Pre-vaccination DENV2 binding reactivity also had no significant correlation with the JEV vaccine response. RVP-based neutralization and enhancement assays against DENV2 showed that there was no significant change in neutralizing or enhancing antibody activity against DENV2 after JEV vaccination.

Conclusions/Significance

This study shows that IMOJEV vaccination elicited a JEV neutralizing response in 97% of vaccinees and that the magnitude of JEV neutralizing titers post-vaccination was not associated with pre-existing binding antibodies to DENV2. Further, while live JEV vaccination increases DENV2-binding antibodies, this cross-reactivity does not lead to DENV2 enhancement. These findings contribute to a better understanding of the orthoflavivirus antibody response following immunization and the influence of pre-existing heterologous orthoflavivirus antibodies. This could guide vaccination strategies, especially in orthoflavivirus-endemic regions.

Author summary

Japanese encephalitis virus (JEV) is the leading pathogen causing viral encephalitis in Asia. Fortunately, there are licensed JEV vaccines incorporated into the routine immunization programs of many Asian countries. In the Philippines, this practice has not yet been adopted on a national scale. This is partly due to concerns over cross-reactive antibody interactions with dengue virus (DENV), a closely related virus that is also endemic in the region. This can happen in a phenomenon known as antibody-dependent enhancement (ADE), which was demonstrated in a previous DENV vaccine trial in the country. To address this, we measured the antibodies against DENV2 and JEV before and after vaccination in children who were administered a live JEV vaccine. We found that most children were able to neutralize JEV post-vaccination, and while this led to increased DENV2 cross-reactivity, the antibodies did not increase DENV2 enhancement in vitro. Our findings suggest that live JEV vaccination is unlikely to increase the risk of severe dengue, at least in the short term. Understanding cross-reactive antibody interactions is crucial for designing vaccines and in shaping vaccine strategies, which are especially relevant in endemic areas.

Introduction

Orthoflaviviruses are a group of single-stranded RNA viruses usually transmitted by arthropod vectors. This virus family includes global public health threats like dengue virus (DENV), Japanese encephalitis virus (JEV), Zika virus (ZIKV), West Nile Virus (WNV), and Yellow fever virus (YFV) [1,2]. DENV, JEV and ZIKV, in particular, are known to be prevalent in tropical countries like the Philippines [35].

Japanese encephalitis virus (JEV), is a virus transmitted by Culex mosquitoes that can cause severe neurologic disease in infected individuals [6]. In Asia, it is the main cause of viral encephalitis [7]. While 99% of individuals infected via mosquito bites remain asymptomatic, the condition is fatal for a third of symptomatic patients. Meanwhile, survivors often deal with long-term neurologic sequelae [6]. Due to the severe nature of the disease, there have been many efforts towards JEV vaccine development since the 1980s. Several vaccines are available, with early ones being inactivated mouse brain-derived or Vero-cell derived vaccines using the Nakayama or Beijing strains. The newer vaccines use the attenuated SA-14-14-2 strain, with the inactivated vaccine mainly used in the United States and Europe, while the live vaccines are more commonly used in Asia [8]. These have been incorporated into the national immunization programs of many Asian countries, such as Japan, China, Korea, Taiwan, and Thailand [9,10]. In the Philippines, JEV is known to be prevalent, so vaccination is recommended by the Philippine Pediatric Society for children at least 9 months of age [11]. However, the vaccine is still not a part of the nationwide routine immunization program [4,10].

DENV, on the other hand, is transmitted by Aedes mosquitoes and is the most prevalent flaviviral illness worldwide, with over 100–400 million infections annually [12]. It causes non-specific flu-like symptoms such as fever, headache, rashes, and joint pains, from which patients typically recover through supportive care. However, in severe cases that usually happen during a secondary DENV infection, more severe hemorrhagic manifestations are observed and may even lead to death [12,13]. Vaccines such as Dengvaxia and Qdenga have been developed for this disease [14], but neither are currently licensed for use in the Philippines.

In this study, we explore two important concepts in orthoflavivirus immunization: the role of pre-existing antibodies and antibody-dependent enhancement. In children, pre-existing orthoflavivirus antibodies may be from maternal antibodies or early natural infection. For other diseases, it is believed that maternal antibodies may interfere with the vaccination response due to immunologic blunting [1519]. It is proposed that this happens when maternal antibodies dampen the infant’s immune response to vaccination through mechanisms such as live virus neutralization or epitope masking [20]. Since the only licensed orthoflavivirus vaccines that are given routinely in childhood are for JEV and YFV [14,21] and because this practice is only done in endemic regions, studies on the influence of maternal antibodies on orthoflavivirus vaccination are limited. Meanwhile, research on heterologous orthoflavivirus infections has shown that, due to the antigenic similarities between these viruses, previous exposure affects the immune response to a subsequent orthoflavivirus encounter. The nature of this effect depends on factors such as the order, timing, and kind (whether natural or vaccine-induced) of exposures; differences may also be found between vaccine types. The existing literature on this topic has been extensively reviewed elsewhere [22,23].

Antibody-dependent enhancement (ADE), on the other hand, is a phenomenon wherein cross-reactive antibodies present at low levels are not capable of neutralizing the virus. In turn, the binding facilitates virus entry and enhancement of infection, which can contribute to severe disease [24]. Health officials in the Philippines have become wary about this due to the country’s previous experience with the Dengvaxia vaccine, wherein clinical trial data for the vaccine showed that children who were DENV-naïve upon immunization had an increased risk of developing severe dengue when exposed to the virus [25]. Parents have also become cautious of vaccination campaigns after the Dengvaxia controversy [2629], and the importance of this phenomenon has initiated its investigation of other existing orthoflavivirus vaccines [30]. For JEV, the current evidence has largely been mixed with key differences in participant recruitment and vaccine regimens between studies, making comparisons difficult [3134]. Further, the previous literature has primarily been focused on inactivated vaccines and on old JEV vaccine strains which are quickly being replaced by live SA-14-14-2 vaccination in Asia [10,35,36].

Given the public health significance of Japanese encephalitis and the need for data to guide vaccination policies in regions that use this vaccine, our group sought to determine if pre-existing anti-DENV2 antibodies would affect the neutralizing titers against JEV post-vaccination and whether immunization with a live attenuated JEV vaccine strain would lead to an increased tendency toward ADE of DENV2 infection in Filipino children. We found that baseline DENV2 binding reactivity was not associated with the JEV vaccine response and observed no significant vaccine-induced DENV2 cross-protection or enhancement in our cohort.

Methods

Ethics statement

The samples used in this work came from a cohort study led by investigators from the University of the Philippines Manila. There were a total of 40 children in the original cohort whose guardians or parents all provided written informed consent. Of the 40 participants, 29 gave consent for future use of biobanked sera. The cohort study was approved by the University of the Philippines Manila Research Ethics Board (UPMREB 2021-0739-01). The use of biobanked sera and access to demographic data for this present study was approved by the Institutional Review Board at the University of Rhode Island (IRB Reference #2225128-1) and the University of the Philippines Manila Research Ethics Board (UPMREB 2024-0613-01).

Serum samples and IMOJEV vaccine

Biobanked sera from 29 children who were 9 months to 2 years old upon vaccination with IMOJEV were used in this study. IMOJEV is a monovalent, chimeric JEV vaccine that uses the yellow fever 17D-204 virus as its backbone, with the premembrane (prM) and envelope (E) sequences replaced by that of the live-attenuated JEV SA-14-14-2 vaccine virus, which was generated from the wild type SA14 strain by serial passaging [37]. Pre- and 1 month-post vaccination samples were available for each child for a total of 58 samples. Recruitment and vaccination were done from July - December 2022 in Tuguegarao City, Cagayan, and Cabagan, Isabela. These are municipalities in the Philippines found in the Cagayan Valley Region, known to be endemic for both JEV and DENV. None of the children who participated in the study received other flavivirus vaccines (i.e., dengue or yellow fever).

DENV2 and JEV VLP IgG ELISA

ELISA assays were performed as previously described [38]. Briefly, clear 96-well flat-bottom plates were coated overnight at 4°C with 10 ng/well of DENV serotype 2 virus-like particle (VLP) or 25 ng/well of JEV VLP (The Native Antigen Company) diluted in coating buffer (Sigma-Aldrich). This was followed by one round of washing using 1X phosphate-buffered saline (PBS) with 0.05% Tween 20 and blocking for 1 hour at 37°C with blocking buffer (Sigma-Aldrich). After blocking, the plates were washed three times. Then, sera diluted to 1:200 in blocking buffer was added to each well and incubated at 37°C for 1 hour. Plates were washed five times before adding goat anti-human IgG with conjugated HRP (Bethyl Lab) diluted 1:25,000 in blocking buffer. This was incubated for 30 minutes at 37°C. After five washes, the detection was done using 1-Step TMB ELISA Substrate Solution (Thermo Scientific). Ten percent 2N sulfuric acid diluted in deionized water was added after 12 minutes to stop the reaction and read at 450 nm and 560 nm. Each serum was assayed in duplicate, and OD450 – OD560 values were averaged. A serum sample known to be negative for DENV IgG (LGC Clinical Diagnostics) was used as a negative control.

Cells

K562 and Raji-DCSIGNR cells were grown in RPMI-1640 Medium, while HEK-293T cells were grown in DMEM, high glucose (HyClone Laboratories Inc.). Both cell culture media were supplemented with 10% heat-inactivated fetal bovine serum (HyClone Laboratories Inc.), 100 U/mL penicillin, and 100 μg/mL streptomycin (Sigma-Aldrich). Cells were maintained at 37°C with 5% CO2.

Generation and titration of reporter virus particles (RVP)

Reporter virus particles were generated by co-transfecting in HEK-293T cells a green fluorescent protein (GFP) tagged West Nile Virus (WNV) replicon plasmid and a structural protein cassette expression plasmid (CprME) for DENV2 strain 16681 (GenBank accession no. NC_001474) or JEV strain 7812474 (GenBank accession nos. EF688633 and U70387) (kind gifts from Dr. Stephen Whitehead and Dr. Gregory Gromowski) as previously described [39,40] with some modifications. Transfection of HEK-293T cells was performed in T75 flasks using 32 μg of DNA (replicon plasmid to CprME plasmid ratio of 1:3) and Lipofectamine 3000 (ThermoFisher Scientific). Cells were incubated for 3 days (DENV2) or 5 days (JEV) at 30°C with 5% CO2 before harvesting the RVPs. Upon harvest, the supernatant was clarified by centrifugation, and aliquots were stored at −80°C prior to use. Titration was performed by adding two-fold serially diluted virus particles to 5 x 104 Raji-DCSIGNR cells and incubating for 42 hours at 37°C. Infection was stopped by adding BD Cytofix Fixation Buffer (BD Biosciences) diluted 1:5 in 1X PBS after two rounds of washing with 1X PBS. The number of GFP-positive cells was measured via flow cytometry using a MACSQuant Analyzer 10 (Miltenyi Biotec), with GFP detected at 488 nm excitation and 525/50nm emission.

RVP neutralization and enhancement assays

Sera were heat-inactivated at 56°C for 30 minutes prior to use. Starting at a dilution of 1:10, three-fold (neutralization) or four-fold (enhancement) serial dilutions of the sera were prepared and incubated for 1 hour at 37°C with an equal volume of solution containing JEV or DENV2 RVP diluted to a concentration yielding <10% infection on titration as described above; the same RVP dilution was used in enhancement assays. The RVP-antibody mixture was then added to 5x104 Raji-DCSIGNR (neutralization) or K562 (enhancement) cells and incubated at 37°C for 42 hours. Fixation and flow cytometry were done as detailed above.

Statistical analyses

For the ELISA assays, positive/negative (P/N) values were calculated by dividing the OD value of each sample by the OD value of the negative control. The cut-off setting was done by identifying all test subjects with a P/N value ≤ 1.0. The standard deviation (SD) for their samples and the negative control was multiplied by three (3SD), and all values above a P/N of 1.0 + 3SD were considered reactive.

Flow cytometry data were analyzed using FlowJo V10.4.2. The cell population of interest was identified and gated using the forward scatter area (FSC-A) and side scatter area (SSC-A) parameters based on cell size and granularity. Gating for positive GFP expression was done using uninfected controls as the baseline. For neutralization assays, the 50% RVP neutralization titer (RVPNT50) was determined by first normalizing the data against medium-only and virus-only controls. With this data, a non-linear regression analysis using the log(inhibitor) vs. normalized response -- variable slope equation was performed. Samples were considered to have neutralizing activity at RVPNT50 ≥ 10. Results were presented as log10 values of the RVPNT50 titers with a value of 5 assigned to titers < 10. The fold enhancement on the antibody-dependent enhancement (ADE) assay was derived by comparing the values with serum in a virus-only control. Samples were considered to have enhancing activity when peak enhancement was ≥ two-fold compared to the control.

Comparisons between groups were done using the Student’s t-test, and correlation analyses were done using the Spearman rank correlation test. Results were considered statistically significant for a p-value < 0.05. All statistical analyses were carried out using GraphPad Prism version 10.3.1.

Results

Baseline DENV2 and JEV binding reactivity in Filipino infants prior to IMOJEV vaccination

To measure the baseline presence of DENV2 antibodies in children prior to vaccination, their binding reactivity was evaluated using a DENV2 VLP IgG ELISA. We compared binding reactivity to DENV2 in children less than or greater than 12 months of age since studies have shown that maternal antibodies would have already waned in most children by this age [41,42]. Of the 29 children, 13 were less than 12 months old, while 16 were 12 months or older upon vaccination. At the set cut-off, 3/13 children from the younger group were considered DENV2-reactive. In contrast, there were 5/16 DENV2-reactive subjects from the older group (Fig 1A), giving an overall DENV2 seroprevalence of 28% that suggests most children in this cohort were DENV2-naïve upon vaccination. There was no significant difference (p = 0.16) between the DENV2 binding reactivities of the two groups. DENV2 binding reactivity also showed a weak non-significant (p = 0.06) positive association (r = 0.36) with age (Fig 1B). The baseline reactivity to JEV VLP was also measured, and 7/8 of the DENV2-reactive subjects were also found to be JEV-reactive (Fig 1C). There was no significant difference in JEV reactivities between groups (p = 0.42), and no significant (p = 0.14) correlation (r = 0.28) was found between JEV reactivity and age (Fig 1D). Further analysis showed a positive correlation between the two assays with a Spearman r of 0.78 (p-value <0.0001) (Fig 1E). The findings suggest that the JEV VLP IgG ELISA is not highly specific for JEV reactivity and instead reflects the binding of cross-reactive DENV2 antibodies.

Fig 1. Baseline IgG binding to DENV2 and JEV VLP.

Fig 1

(A and C) Binding reactivity to (A) DENV2 or (C) JEV of children younger than 12 months versus children 12 months and older prior to vaccination (two-tailed Student’s t-test; broken lines at P/N cut-off; horizontal bars represent the mean). (B and D) Relationship between age and binding reactivity against (B) DENV2 or (D) JEV (two-tailed Spearman rank correlation test). (E) Relationship between DENV2 and JEV binding reactivities (two-tailed Spearman rank correlation test).

JEV binding reactivity and neutralization after IMOJEV vaccination

Next, vaccine response was characterized through an RVP neutralization assay against JEV. None of the children showed neutralization activity against JEV prior to vaccination (S1 Fig). At 1 month post-vaccination, there was a significant increase in JEV neutralizing antibody titer in a majority of the children (p < 0.0001), with 28/29 (97%) having RVPNT50 titers ≥ 10 (geometric mean titer: 254.7) (Fig 2A). Only subject B27 failed to respond to vaccination. The JEV IgG binding reactivity also showed a significant increase compared to baseline (p = 0.001) (Fig 2B). However, the ELISA was only able to detect JEV reactivity in half (14/28) of the vaccinees, showing the poor sensitivity of this assay for vaccine-induced antibodies at 1 month post-vaccination. Additionally, 7 out of the 14 subjects were already JEV-reactive pre-vaccination despite having no JEV-neutralizing activity, and an increase in reactivity to JEV was also seen for subject B27 despite the lack of a neutralizing antibody response. Four subjects (B12, B24, B37, B40) were noted to have decreased ELISA reactivity to JEV post-vaccination.

Fig 2. Antibody response to IMOJEV vaccination.

Fig 2

(A) Comparison of log10 JEV RVPNT50 titers pre- and post-vaccination (one-tailed paired Student’s t-test; titers less than 10 were assigned a value of 5). (B) Pairwise comparison of JEV VLP IgG binding reactivity pre- and post-vaccination (one-tailed paired Student’s t-test; in red is subject B27, who did not respond to vaccination based on the RVP neutralization assay). (C and D) Comparison of post-vaccination log10 JEV RVPNT50 titers (C) between age groups and (D) by DENV2 reactivity (two-tailed unpaired Student’s t-test).

We also explored whether the magnitude of vaccine response was associated with age or pre-vaccination DENV2 binding reactivity. We found no significant differences between age groups (Figs 2C and S2A; p = 0.35 and 0.24, respectively). The geometric mean titer of JEV-neutralizing antibodies post-vaccination was lower for the DENV2-reactive group than the non-reactive group (324 vs. 135.3, respectively), but this difference was not statistically significant (p = 0.11, Fig 2D). Pre-vaccination DENV2 binding reactivity also had a weak non-significant (p = 0.09) negative correlation (r = -0.32) with JEV titers post-vaccination (S2B Fig).

DENV2 binding reactivity and functional antibody responses after IMOJEV vaccination

Next, antibody responses to DENV2 were evaluated in 17 subjects that were selected based on age, with 9 from the younger (<12 months) age group and 8 from the older (≥12 months) age group. Among these 17 children, 7 were DENV2-reactive at baseline, while 10 were not. Fig 3A shows that, like JEV VLP binding reactivity, DENV2 VLP binding significantly increased 1 month after immunization in most subjects (p = 0.003). Six children (B12, B24, B38, B35, B37, B39) had decreased reactivities post-vaccination. Consistent with the previous observation on the relationship between DENV2 and JEV ELISA results, the fold change in P/N values after vaccination was also positively correlated between the two assays with a Spearman r of 0.62 (p = 0.0002) (Fig 3B).

Fig 3. DENV2 binding reactivity and functional antibody responses.

Fig 3

(A) Pairwise comparison of DENV2 VLP IgG binding reactivity pre- and post-vaccination (one-tailed paired Student’s t-test). (B) Relationship between the fold change in DENV2 and JEV VLP IgG ELISA P/N values after vaccination (one-tailed Spearman rank correlation test). (C and D) Pairwise comparison of DENV2 neutralizing titers or fold enhancement post-JEV immunization (two-tailed paired Student’s t-test; broken lines at cut-off for neutralization or enhancement and shown in red circles is subject B27 who did not respond to JEV vaccination; in Fig 3C, the line at y = 0.7 represents 13 subjects that did not have neutralizing activity pre- and post-vaccination).

To determine if the DENV2-reactive antibodies in these children are also neutralizing, an RVP neutralization assay against DENV2 was performed. The DENV2 serotype was chosen as the focus of this study due to its prevalence in the Philippines and its relevance to ADE [43,44]. ELISA assays against DENV 1, 3 and 4 VLP antigens also showed a high degree of correlation (Pearson r = 0.66 to 0.78) (S3 Fig) with DENV2 antibody binding. In this set, only 4 out of 7 DENV2-reactive subjects showed neutralization against DENV2 post-vaccination (S4 Fig). Three of them (B01, B10, B27) also showed pre-vaccination neutralizing activity against DENV2 (Fig 3C); all were from the older age group, which further supports that these antibodies are likely from natural DENV exposure. Of the four children, three were vaccine responders (B01, B10, B24), and their DENV2 neutralization titers increased slightly post-JEV immunization (Fig 3C). Subject B27, who did not develop JEV-neutralizing antibodies, showed a decline in DENV2-neutralizing antibodies after 1 month.

Since half of the DENV2-reactive children did not show DENV2 neutralization, we hypothesized that the observed binding is from subneutralizing antibodies that may enhance DENV infection. We also wanted to know whether JEV vaccination would increase any observed enhancement compared to pre-vaccination sera. To evaluate this, an in vitro DENV2 RVP ADE assay in K562 cells was performed. Six out of the 7 DENV2-reactive children also enhanced DENV2 infection in vitro (S5 Fig). However, no significant change in enhancement activity was found post-vaccination (p = 0.27) (Fig 3D and Table 1). Subjects with enhancing but non-neutralizing antibodies to DENV2 (E + /N-) prior to vaccination tended to have the highest neutralizing antibody responses to JEV post-vaccination, but the differences were not statistically significant (S6 Fig). A master table of the ELISA, neutralization and enhancement assay results can be seen in S1 Table.

Table 1. Serum dilution at peak fold enhancement of DENV2.

Participant ID Pre-vaccination Post-vaccination
B01 1:160 1:160
B06 1:10 1:40
B10 1:640 1:640
B12 1:10 1:10
B15 1:10 1:10
B19 1:40 1:10
B22 1:40 1:10
B23 1:10 1:10
B24 1:10 1:10
B25 1:10 1:10
B27 1:160 1:640
B31 1:10 1:40
B32 1:40 1:40
B34 1:40 1:40
B36 1:10 1:10
B37 1:10 1:10
B38 1:10 1:10

Discussion

Reports of severe dengue after DENV vaccination have led to increased vaccine hesitancy among parents in the Philippines [2629]. This may be one of the reasons why earlier efforts to include JEV in the national immunization program have since stalled. Meanwhile, cases of JEV continue to plague the country, with an average annual incidence rate of 1 per million total population since 2008, according to the World Health Organization (WHO) [45]. This is much higher than neighboring countries with routine immunization programs, such as Japan (0.03), South Korea (0.3) and Thailand (0.45). Such statistics underscore the need for routine JEV immunization for Filipino children. Here, we report that children from a JEV- and DENV-endemic region in the Philippines had strong responses to JEV vaccination regardless of their pre-existing DENV2 binding antibodies and that vaccination elicited cross-reactive antibodies to DENV2, but these did not enhance DENV2 infection in vitro.

In this cohort, we found a baseline DENV2 seroprevalence of 28%. The lower seropositivity rate in our study is consistent with the observation that DENV seropositivity tends to increase in older age groups [46] and stresses the burden of orthoflavivirus exposure in the region. We also measured the baseline IgG seroprevalence against JEV using ELISA but found that the results reflected DENV cross-reactivity rather than true JEV binding, highlighting the poor performance of this assay in detecting JEV vaccine response similar to what other groups have observed [4749]. Our preliminary testing on a subset of samples also showed that vaccine response could not be detected at antigen concentrations of 10, 25 and 50 ng/well (S2 Table). A correlation matrix between the DENV2 and JEV ELISA binding reactivity values with neutralization titers also showed that DENV2 correlation is high for both pre- and post-vaccination samples (Pearson r = 0.74 to 0.76), but this is not the case for JEV (S7 Fig). The presence of vaccine-induced anti-JEV IgM in the serum may also explain the findings. In previous studies, it was shown that 30–40% of vaccinees still had detectable IgM levels one month post-vaccination which may interfere with IgG detection [49,50].

Analyzing the relationship between DENV2 reactivities and age, we found a positive correlation between the two. In older children, this likely reflects natural DENV exposure rather than maternal DENV antibodies as supported by finding DENV neutralizing antibodies only in older children. However, the weak and non-significant correlation also implies that maternal antibodies may still be present in some children. In infants, DENV antibodies are usually expected to decrease after birth due to the waning of maternal antibodies, but these may already be undetectable in older infants [41,42,51].

The neutralization and ADE assays were performed using an RVP-based platform previously developed by others [39,40]. The use of RVPs allows for high-throughput and rapid evaluation of functional antibody responses to viruses while being relatively safer compared to the traditional plaque assay due to the non-replicating nature of the pseudoinfectious particles [52,53]. In this assay, RVP infection of cells is measured via the optical detection of a GFP-tagged reporter gene expressed by the RVPs [40,52]. For JEV, neutralizing titers ≥ 10 on a plaque reduction neutralization test (PRNT) are considered seroprotective [54]. In this study, 97% of the children developed neutralizing titers ≥ 10 against JEV 28 days after vaccination, which is indicative of a good vaccine response, as previous groups have reported for the IMOJEV vaccine [55,56]. We note, however, that the antibody level threshold for the RVP-based neutralization assay has not yet been validated. We also examined JEV vaccine response by age to explore if maternal antibodies, which may still be present in some younger children, would interfere with the immune response. This a phenomenon that has been observed in vaccines for diseases such as pertussis, measles and malaria [1719]. In our study, JEV vaccine responses did not differ by age and pre-vaccination DENV2 binding reactivity was not shown to be associated with vaccine response. However, children with pre-existing DENV2 enhancing and neutralizing antibodies showed lower vaccine responses. This may be reflective of original antigenic sin, where immunologic memory blunts the immune response to a similar but non-identical antigen [57], as also previously observed for the YFV vaccine [58]. Notably, children with enhancing but non-neutralizing DENV2 antibodies had higher JEV vaccine responses, consistent with a hypothesis that cross-reactive maternal DENV antibodies may boost the immune response through enhancement of the live attenuated virus [59].

Another significant concern in orthoflavivirus vaccine response that has not yet been well-explored for this vaccine is antibody-dependent enhancement, which is a phenomenon that makes vaccine design for orthoflaviviruses challenging since cross-reactive antibodies could be neutralizing but at certain conditions, may also be enhancing [22]. Besides Dengvaxia, other orthoflavivirus vaccines have also demonstrated cross-reactive binding to related viruses, with studies showing that this could lead to both cross-protection and enhancement [30,60,61]. In this study, we observed no significant change in DENV2-enhancing activity post-vaccination.

Other literature exploring the effects of JEV antibodies on DENV enhancement has been limited, and available studies have mixed findings. A cohort from Thailand recruited between 1998–2002 observed that the presence of detectable JEV antibodies was associated with an increased risk of symptomatic DENV infection [31]. Another study of Japanese adults vaccinated from 2009-2011 also revealed that more post-vaccination subjects exhibited in vitro DENV ADE compared to pre-vaccination subjects [32]. However, a third study conducted in adults from Thailand recruited in 1982 concluded that pre-existing JEV antibodies did not enhance DENV2 infection [33]. Notably, the third study involved a cohort that likely developed antibodies from natural exposure since recruitment occurred prior to routine immunization in the country. Moreover, a JEV vaccine trial conducted in 1984–1985 involving a cohort of over 65,000 children in Thailand found that there was a lower number of dengue fever and severe dengue cases among children from the vaccinated group, a trend that was observed until two years post-vaccination [34]. While they did not explore enhancement, the authors state that this finding points to possible DENV cross-protection from JEV vaccination, especially since the difference in DENV attack rates between groups was highest in the months following vaccination. In the present study, live vaccination using a chimeric JEV SA-14-14-2 strain was given to children, while the previous vaccine studies used cohorts that were given JEVAX (inactivated mouse brain-derived vaccine using Nakayama strain), JEBIK-V (inactivated Vero cell vaccine using Beijing-1 strain) and in the case of the study by Hoke et al. (1988), an inactivated monovalent or bivalent vaccine containing either the Nakayama strain alone or in combination with the Beijing-1 strain. Potential differences in enhancing activity by various JEV strains were proposed by a recent paper that identified the 106th and 107th amino acids on the envelope (E) protein as ADE-inducing epitopes. Mutating these amino acids in Nakayama, Beijing-1, and P3 strains of JEV lowered the observed DENV enhancement in immunized mice [62]. Interestingly, the L107F mutation they induced in these strains is already found on the SA-14-14-2 E protein. Differences in the mode of vaccine delivery may also play a role. Still, an in vivo study in mice found that both inactivated and live attenuated JEV vaccines can cross-protect against DENV 1–4 with no increased mortality indicative of enhancement [63]. Other key differences that may influence study findings include JEV exposure (whether natural or from immunization), cohort age, and the recruitment region and time period.

Our study has several limitations that should be considered when interpreting these results. First, the assays on DENV were focused on one serotype and strain (DENV 2 16681). It is possible that cross-reactive antibodies may behave differently against other dengue viruses. We were also limited by the sample size of our cohort. Similar studies on other vaccination cohorts would be useful to validate the results of this study. Being a live vaccine, IMOJEV is recommended as a one-dose regimen, but booster doses may also be given, and our study did not characterize the effect of such follow-up immunization. Further, since only one-month post-vaccination samples were tested, we cannot draw firm conclusions about the antibody responses at later time points. In this endemic population, such studies would also be confounded by orthoflavivirus exposures in the interim. Lastly, while previous studies have shown an association between in vitro enhancement and severe secondary dengue illness [62,64], it is important to recognize the limitations in interpreting ADE assay results and their clinical implications. More studies using other ADE models or prospective cohorts observing vaccinated children are recommended.

Japanese encephalitis virus continues to be endemic in parts of Asia, but the resulting disease is preventable through vaccination. In this study, we show that IMOJEV vaccination can elicit neutralizing titers against JEV in Filipino children and that this immunization does not result in increased in vitro enhancement of DENV2 infection. We believe the results support routine immunization, especially in children who live in known JEV hotspots. As orthoflaviviruses continue to circulate worldwide, concurrent with the rollout of new vaccines and routine immunization for existing vaccines, more investigations on the orthoflavivirus immune response after natural exposure and vaccination are needed, as well as on the effect of heterologous exposures on these antibody responses.

Supporting information

S1 Fig. Pre- and post- IMOJEV vaccination anti-JEV neutralizing titers.

(TIF)

pntd.0013550.s001.tif (969.4KB, tif)
S2 Fig. Relationship between the log10 JEV RVPNT50 titers and (A) age or (B) DENV2 binding reactivities (two-tailed Spearman rank correlation test).

(TIF)

pntd.0013550.s002.tif (76.1KB, tif)
S3 Fig. Correlation between DENV2 ELISA P/N Values and DENV 1, 3 and 4 ELISA P/N Values.

(TIF)

pntd.0013550.s003.tif (467.7KB, tif)
S4 Fig. Pre- and post-IMOJEV vaccination anti-DENV2 neutralizing titers.

(TIF)

pntd.0013550.s004.tif (471.9KB, tif)
S5 Fig. Pre- and post-IMOJEV vaccination DENV2 antibody dependent enhancement of selected samples.

(TIF)

pntd.0013550.s005.tif (314.9KB, tif)
S6 Fig. Comparison of vaccine response based on pre-IMOJEV vaccination DENV2 functional antibody response.

(TIF)

pntd.0013550.s006.tif (39.7KB, tif)
S7 Fig. Correlation between (A) DENV2 or (B) JEV ELISA P/N values with neutralization titers.

(TIF)

pntd.0013550.s007.tif (623.7KB, tif)
S1 Table. Master table of ELISA, neutralization and enhancement assay results.

(XLSX)

pntd.0013550.s008.xlsx (12.6KB, xlsx)
S2 Table. Test of different JEV antigen concentrations for selected serum samples at 1:200 serum dilution.

(XLSX)

pntd.0013550.s009.xlsx (9.4KB, xlsx)

Acknowledgments

We wish to thank the Department of Science and Technology - Philippine Council for Health Research and Development (DOST-PCHRD) for SLMD’s MECO-TECO JRP research funding, which supported the cohort study that provided samples for this study and the National Institutes of Health (NIH) for ALR’s grant funding (U01 AI179523), which provided support for the in vitro experiments. We thank Dr. Stephen Whitehead (US National Institutes of Health) and Dr. Gregory Gromowski (Walter Reed Army Institute of Research) for generously sharing their plasmids for use in this study as well as Dr. Day-Yu Chao for her intellectual contributions to the parent study from which this work arose. We also recognize Mr. Scott de Sagon for his technical support, and Ms. Calline Danica Gomez, Ms. Kiara Maye Sta. Ana, Ms. Edelyn Ibe and Mr. John Clarence Flores for their administrative assistance. Finally, we gratefully acknowledge the children who participated in the cohort study and their parents for providing consent.

Data Availability

Data Availability Statement: All relevant data are within the manuscript and its Supporting information files, with the exception of exact participant age, which is potentially identifying. Access to this data is restricted by the University of the Philippines Manila Research Ethics Board (UPMREB) to protect participant confidentiality. Data may be made available upon reasonable request to the UPMREB (upmreb@post.upm.edu.ph).

Funding Statement

This project was done as part of FESV’s Fulbright fellowship. We gratefully acknowledge the support of the Fulbright U.S. Student Program, which is sponsored by the U.S. Department of State and the Philippine-American Educational Foundation (PAEF). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The contents are solely the responsibility of the authors and do not necessarily represent the official views of the Fulbright Program, the U.S. Department of State, or PAEF.

References

  • 1.Lindenbach BD, Murray CL, Thiel H-J, Rice CM. Flaviviridae. In: Knipe DM, Howley PM, editors. Fields Virology. 6th ed. Philadelphia, PA: Wolters Kluwer/Lippincott Williams & Wilkins Health; 2013. p. 712–46. [Google Scholar]
  • 2.Gaunt MW, Sall AA, Lamballerie X de, Falconar AKI, Dzhivanian TI, Gould EA. Phylogenetic relationships of flaviviruses correlate with their epidemiology, disease association and biogeography. J Gen Virol. 2001;82(Pt 8):1867–76. doi: 10.1099/0022-1317-82-8-1867 [DOI] [PubMed] [Google Scholar]
  • 3.Edillo FE, Halasa YA, Largo FM, Erasmo JNV, Amoin NB, Alera MTP, et al. Economic cost and burden of dengue in the Philippines. Am J Trop Med Hyg. 2015;92(2):360–6. doi: 10.4269/ajtmh.14-0139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lopez AL, Aldaba JG, Roque VG Jr, Tandoc AO 3rd, Sy AK, Espino FE, et al. Epidemiology of Japanese encephalitis in the Philippines: a systematic review. PLoS Negl Trop Dis. 2015;9(3):e0003630. doi: 10.1371/journal.pntd.0003630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Biggs JR, Sy AK, Brady OJ, Kucharski AJ, Funk S, Tu Y-H, et al. Serological Evidence of Widespread Zika Transmission across the Philippines. Viruses. 2021;13(8):1441. doi: 10.3390/v13081441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sharma KB, Vrati S, Kalia M. Pathobiology of Japanese encephalitis virus infection. Mol Aspects Med. 2021;81:100994. doi: 10.1016/j.mam.2021.100994 [DOI] [PubMed] [Google Scholar]
  • 7.Monath TP. Japanese Encephalitis: Risk of Emergence in the United States and the Resulting Impact. Viruses. 2023;16(1):54. doi: 10.3390/v16010054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Asawapaithulsert P, Ngamprasertchai T, Kitro A. Japanese Encephalitis Vaccine Acceptance and Strategies for Travelers: Insights from a Scoping Review and Practitioners in Endemic Countries. Vaccines (Basel). 2023;11(11):1683. doi: 10.3390/vaccines11111683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen H-L, Chang J-K, Tang R-B. Current recommendations for the Japanese encephalitis vaccine. J Chin Med Assoc. 2015;78(5):271–5. doi: 10.1016/j.jcma.2014.12.009 [DOI] [PubMed] [Google Scholar]
  • 10.Heffelfinger JD, Li X, Batmunkh N, Grabovac V, Diorditsa S, Liyanage JB, et al. Japanese Encephalitis Surveillance and Immunization - Asia and Western Pacific Regions, 2016. MMWR Morb Mortal Wkly Rep. 2017;66(22):579–83. doi: 10.15585/mmwr.mm6622a3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Philippine Infectious Disease Society of the Philippines, Philippine Pediatric Society, Philippine Foundation for Vaccination. Childhood Immunization Schedule 2024. 2024. Available from: https://www.pidsphil.org/home/wp-content/uploads/2024/03/CHILDHOOD-IMMUNIZATION-SCHEDULE-2024.pdf
  • 12.World Health Organization. Dengue and severe dengue. 23 Apr 2024 [cited 1 Dec 2024]. Available from: https://www.who.int/news-room/fact-sheets/detail/dengue-and-severe-dengue
  • 13.Guzman MG, Gubler DJ, Izquierdo A, Martinez E, Halstead SB. Dengue infection. Nat Rev Dis Primers. 2016;2:16055. doi: 10.1038/nrdp.2016.55 [DOI] [PubMed] [Google Scholar]
  • 14.Staples JE. Flavivirus vaccines: policy and market considerations. Centers for Disease Control and Prevention; 20 Apr 2023. Available from: https://cdn.who.int/media/docs/default-source/immunization/mrna-ttp/april-2023/3_staples_flavis_policy.pdf?sfvrsn=f3b93e8d_2 [Google Scholar]
  • 15.Cinicola B, Conti MG, Terrin G, Sgrulletti M, Elfeky R, Carsetti R, et al. The Protective Role of Maternal Immunization in Early Life. Front Pediatr. 2021;9:638871. doi: 10.3389/fped.2021.638871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Vono M, Eberhardt CS, Auderset F, Mastelic-Gavillet B, Lemeille S, Christensen D, et al. Maternal Antibodies Inhibit Neonatal and Infant Responses to Vaccination by Shaping the Early-Life B Cell Repertoire within Germinal Centers. Cell Rep. 2019;28(7):1773-1784.e5. doi: 10.1016/j.celrep.2019.07.047 [DOI] [PubMed] [Google Scholar]
  • 17.Maertens K, Hoang TTH, Nguyen TD, Caboré RN, Duong TH, Huygen K, et al. The Effect of Maternal Pertussis Immunization on Infant Vaccine Responses to a Booster Pertussis-Containing Vaccine in Vietnam. Clin Infect Dis. 2016;63(suppl 4):S197–204. doi: 10.1093/cid/ciw551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nic Lochlainn LM, de Gier B, van der Maas N, van Binnendijk R, Strebel PM, Goodman T, et al. Effect of measles vaccination in infants younger than 9 months on the immune response to subsequent measles vaccine doses: a systematic review and meta-analysis. Lancet Infect Dis. 2019;19(11):1246–54. doi: 10.1016/S1473-3099(19)30396-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Macià D, Campo JJ, Jairoce C, Mpina M, Sorgho H, Dosoo D, et al. The effect of Plasmodium falciparum exposure and maternal anti-circumsporozoite protein antibodies on responses to RTS,S/AS01E vaccination in infants and children: an ancillary observational immunological study to a phase 3, randomised clinical trial. Lancet Infect Dis. 2025;25(3):335–45. doi: 10.1016/S1473-3099(24)00527-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Siegrist CA. Mechanisms by which maternal antibodies influence infant vaccine responses: review of hypotheses and definition of main determinants. Vaccine. 2003;21(24):3406–12. doi: 10.1016/s0264-410x(03)00342-6 [DOI] [PubMed] [Google Scholar]
  • 21.Gavi The Vaccine Alliance. Yellow fever vaccine support. In: Gavi The Vaccine Alliance [Internet]. 18 Nov 2024 [cited 1 Dec 2024]. Available from: https://www.gavi.org/types-support/vaccine-support/yellow-fever
  • 22.Rathore APS, St John AL. Cross-Reactive Immunity Among Flaviviruses. Front Immunol. 2020;11:334. doi: 10.3389/fimmu.2020.00334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rey FA, Stiasny K, Vaney M-C, Dellarole M, Heinz FX. The bright and the dark side of human antibody responses to flaviviruses: lessons for vaccine design. EMBO Rep. 2018;19(2):206–24. doi: 10.15252/embr.201745302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Halstead SB. Dengue Antibody-Dependent Enhancement: Knowns and Unknowns. Microbiol Spectr. 2014;2(6). doi: 10.1128/microbiolspec.AID-0022-2014 [DOI] [PubMed] [Google Scholar]
  • 25.Halstead SB. Dengvaxia sensitizes seronegatives to vaccine enhanced disease regardless of age. Vaccine. 2017;35(47):6355–8. doi: 10.1016/j.vaccine.2017.09.089 [DOI] [PubMed] [Google Scholar]
  • 26.Miras AP, Regencia ZJG, Baja ES. “I was terrified for my child”: understanding the link between the Dengvaxia® controversy and the measles vaccine hesitancy in Pasay City, Philippines. J Public Health (Oxf). 2023;45(4):912–8. doi: 10.1093/pubmed/fdad091 [DOI] [PubMed] [Google Scholar]
  • 27.Yu VG, Lasco G, David CC. Fear, mistrust, and vaccine hesitancy: Narratives of the dengue vaccine controversy in the Philippines. Vaccine. 2021;39(35):4964–72. doi: 10.1016/j.vaccine.2021.07.051 [DOI] [PubMed] [Google Scholar]
  • 28.Mabale MAA, Tejero LMS, Montes LA, Collante MTM, Tempongko MSB, Tolabing MCC. Implications of information heard about Dengvaxia on Filipinos’ perception on vaccination. Vaccine. 2024;42(7):1673–81. doi: 10.1016/j.vaccine.2024.01.097 [DOI] [PubMed] [Google Scholar]
  • 29.Larson HJ, Hartigan-Go K, de Figueiredo A. Vaccine confidence plummets in the Philippines following dengue vaccine scare: why it matters to pandemic preparedness. Hum Vaccin Immunother. 2019;15(3):625–7. doi: 10.1080/21645515.2018.1522468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Santos-Peral A, Luppa F, Goresch S, Nikolova E, Zaucha M, Lehmann L, et al. Prior flavivirus immunity skews the yellow fever vaccine response to cross-reactive antibodies with potential to enhance dengue virus infection. Nat Commun. 2024;15(1):1696. doi: 10.1038/s41467-024-45806-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Anderson KB, Gibbons RV, Thomas SJ, Rothman AL, Nisalak A, Berkelman RL, et al. Preexisting Japanese encephalitis virus neutralizing antibodies and increased symptomatic dengue illness in a school-based cohort in Thailand. PLoS Negl Trop Dis. 2011;5(10):e1311. doi: 10.1371/journal.pntd.0001311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Saito Y, Moi ML, Takeshita N, Lim C-K, Shiba H, Hosono K, et al. Japanese encephalitis vaccine-facilitated dengue virus infection-enhancement antibody in adults. BMC Infect Dis. 2016;16(1):578. doi: 10.1186/s12879-016-1873-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Putvatana R, Yoksan S, Chayayodhin T, Bhamarapravati N, Halstead SB. Absence of dengue 2 infection enhancement in human sera containing Japanese encephalitis antibodies. Am J Trop Med Hyg. 1984;33(2):288–94. doi: 10.4269/ajtmh.1984.33.288 [DOI] [PubMed] [Google Scholar]
  • 34.Hoke CH, Nisalak A, Sangawhipa N, Jatanasen S, Laorakapongse T, Innis BL, et al. Protection against Japanese encephalitis by inactivated vaccines. N Engl J Med. 1988;319(10):608–14. doi: 10.1056/NEJM198809083191004 [DOI] [PubMed] [Google Scholar]
  • 35.Hsu J-Y, Hung C-C, Tsou T-P, Chen W-C. Epidemiology and risk factors of Japanese encephalitis in Taiwan, 2010-2022. PLoS Negl Trop Dis. 2023;17(10):e0011421. doi: 10.1371/journal.pntd.0011421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sudjaritruk T, Kaewpoowat Q, Prasarakee C, Sarachai S, Taurel A-F, Sricharoen N, et al. Seroepidemiological study of Japanese encephalitis virus in Chiang Mai: Immunity and susceptibility 28 years after introduction of a vaccination programme. PLoS Negl Trop Dis. 2022;16(8):e0010674. doi: 10.1371/journal.pntd.0010674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Appaiahgari MB, Vrati S. IMOJEV(®): a Yellow fever virus-based novel Japanese encephalitis vaccine. Expert Rev Vaccines. 2010;9(12):1371–84. doi: 10.1586/erv.10.139 [DOI] [PubMed] [Google Scholar]
  • 38.Sanchez-Vargas LA, Mathew A, Salje H, Sousa D, Casale NA, Farmer A, et al. Protective Role of NS1-Specific Antibodies in the Immune Response to Dengue Virus Through Antibody-Dependent Cellular Cytotoxicity. J Infect Dis. 2024;230(5):1147–56. doi: 10.1093/infdis/jiae137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pierson TC, Xu Q, Nelson S, Oliphant T, Nybakken GE, Fremont DH, et al. The stoichiometry of antibody-mediated neutralization and enhancement of West Nile virus infection. Cell Host Microbe. 2007;1(2):135–45. doi: 10.1016/j.chom.2007.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mukherjee S, Pierson TC, Dowd KA. Pseudo-infectious Reporter Virus Particles for Measuring Antibody-Mediated Neutralization and Enhancement of Dengue Virus Infection. In: Padmanabhan R, Vasudevan SG, editors. Dengue. New York, NY: Springer New York; 2014. p. 75–97. doi: 10.1007/978-1-4939-0348-1_6 [DOI] [PubMed] [Google Scholar]
  • 41.Simmons CP, Chau TNB, Thuy TT, Tuan NM, Hoang DM, Thien NT, et al. Maternal antibody and viral factors in the pathogenesis of dengue virus in infants. J Infect Dis. 2007;196(3):416–24. doi: 10.1086/519170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.van Panhuis WG, Luxemburger C, Pengsaa K, Limkittikul K, Sabchareon A, Lang J, et al. Decay and persistence of maternal dengue antibodies among infants in Bangkok. Am J Trop Med Hyg. 2011;85(2):355–62. doi: 10.4269/ajtmh.2011.11-0125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Guzman MG, Vazquez S. The complexity of antibody-dependent enhancement of dengue virus infection. Viruses. 2010;2(12):2649–62. doi: 10.3390/v2122649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Balingit JC, Dimamay MPS, Suzuki R, Matsuda M, Xayavong D, Ngwe Tun MM, et al. Role of pre-existing immunity in driving the dengue virus serotype 2 genotype shift in the Philippines: A retrospective analysis of serological data. Int J Infect Dis. 2024;139:59–68. doi: 10.1016/j.ijid.2023.11.025 [DOI] [PubMed] [Google Scholar]
  • 45.Japanese Encephalitis (JE) reported cases and incidence. In: Japanese Encephalitis (JE) reported cases and incidence [Internet]. [cited 11 Nov 2024]. Available from: https://immunizationdata.who.int/global/wiise-detail-page
  • 46.Alera MT, Srikiatkhachorn A, Velasco JM, Tac-An IA, Lago CB, Clapham HE, et al. Incidence of Dengue Virus Infection in Adults and Children in a Prospective Longitudinal Cohort in the Philippines. PLoS Negl Trop Dis. 2016;10(2):e0004337. doi: 10.1371/journal.pntd.0004337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kinsella P, Moso M, Martin G, Karapangiotidis T, Karamalakis D, Nicholson S, et al. Laboratory evaluation of ELISA and indirect immunofluorescence assay in response to emergence of Japanese encephalitis virus genotype IV in Australia. J Clin Virol. 2023;168:105580. doi: 10.1016/j.jcv.2023.105580 [DOI] [PubMed] [Google Scholar]
  • 48.Cha GW, Cho JE, Ju YR, Hong Y-J, Han MG, Lee W-J, et al. Comparison of four serological tests for detecting antibodies to Japanese encephalitis virus after vaccination in children. Osong Public Health Res Perspect. 2014;5(5):286–91. doi: 10.1016/j.phrp.2014.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Litzba N, Klade CS, Lederer S, Niedrig M. Evaluation of serological diagnostic test systems assessing the immune response to Japanese encephalitis vaccination. PLoS Negl Trop Dis. 2010;4(11):e883. doi: 10.1371/journal.pntd.0000883 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hills SL, Van Keulen A, Feser J, Panella A, Letson GW, Staples JE, et al. Persistence of IgM Antibodies after Vaccination with Live Attenuated Japanese Encephalitis Vaccine. Am J Trop Med Hyg. 2020;104(2):576–9. doi: 10.4269/ajtmh.20-1132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.O’Driscoll M, Buddhari D, Huang AT, Waickman A, Kaewhirun S, Iamsirithaworn S, et al. Maternally derived antibody titer dynamics and risk of hospitalized infant dengue disease. Proc Natl Acad Sci U S A. 2023;120(41):e2308221120. doi: 10.1073/pnas.2308221120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mattia K, Puffer BA, Williams KL, Gonzalez R, Murray M, Sluzas E, et al. Dengue reporter virus particles for measuring neutralizing antibodies against each of the four dengue serotypes. PLoS One. 2011;6(11):e27252. doi: 10.1371/journal.pone.0027252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lingemann M, Amaro-Carambot E, Lamirande EW, Pierson TC, Whitehead SS. Simultaneous quantitation of neutralizing antibodies against all four dengue virus serotypes using optimized reporter virus particles. J Virol. 2024;98(7):e0068124. doi: 10.1128/jvi.00681-24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hombach J, Solomon T, Kurane I, Jacobson J, Wood D. Report on a WHO consultation on immunological endpoints for evaluation of new Japanese encephalitis vaccines, WHO, Geneva, 2-3 September, 2004. Vaccine. 2005;23(45):5205–11. doi: 10.1016/j.vaccine.2005.07.002 [DOI] [PubMed] [Google Scholar]
  • 55.Chokephaibulkit K, Houillon G, Feroldi E, Bouckenooghe A. Safety and immunogenicity of a live attenuated Japanese encephalitis chimeric virus vaccine (IMOJEV®) in children. Expert Rev Vaccines. 2016;15(2):153–66. doi: 10.1586/14760584.2016.1123097 [DOI] [PubMed] [Google Scholar]
  • 56.Vu TD, Nguyen QD, Tran HTA, Bosch-Castells V, Zocchetti C, Houillon G. Immunogenicity and safety of a single dose of a live attenuated Japanese encephalitis chimeric virus vaccine in Vietnam: A single-arm, single-center study. Int J Infect Dis. 2018;66:137–42. doi: 10.1016/j.ijid.2017.10.010 [DOI] [PubMed] [Google Scholar]
  • 57.Vatti A, Monsalve DM, Pacheco Y, Chang C, Anaya J-M, Gershwin ME. Original antigenic sin: A comprehensive review. J Autoimmun. 2017;83:12–21. doi: 10.1016/j.jaut.2017.04.008 [DOI] [PubMed] [Google Scholar]
  • 58.Bradt V, Malafa S, von Braun A, Jarmer J, Tsouchnikas G, Medits I, et al. Pre-existing yellow fever immunity impairs and modulates the antibody response to tick-borne encephalitis vaccination. NPJ Vaccines. 2019;4:38. doi: 10.1038/s41541-019-0133-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mok DZL, Chan KR. The Effects of Pre-Existing Antibodies on Live-Attenuated Viral Vaccines. Viruses. 2020;12(5):520. doi: 10.3390/v12050520 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yamshchikov G, Borisevich V, Kwok CW, Nistler R, Kohlmeier J, Seregin A, et al. The suitability of yellow fever and Japanese encephalitis vaccines for immunization against West Nile virus. Vaccine. 2005;23(39):4785–92. doi: 10.1016/j.vaccine.2005.04.036 [DOI] [PubMed] [Google Scholar]
  • 61.Orlinger KK, Hofmeister Y, Fritz R, Holzer GW, Falkner FG, Unger B, et al. A tick-borne encephalitis virus vaccine based on the European prototype strain induces broadly reactive cross-neutralizing antibodies in humans. J Infect Dis. 2011;203(11):1556–64. doi: 10.1093/infdis/jir122 [DOI] [PubMed] [Google Scholar]
  • 62.Kotaki T, Nagai Y, Yamanaka A, Konishi E, Kameoka M. Japanese Encephalitis DNA Vaccines with Epitope Modification Reduce the Induction of Cross-Reactive Antibodies against Dengue Virus and Antibody-Dependent Enhancement of Dengue Virus Infection. Vaccines (Basel). 2022;10(9):1411. doi: 10.3390/vaccines10091411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li J, Gao N, Fan D, Chen H, Sheng Z, Fu S, et al. Cross-protection induced by Japanese encephalitis vaccines against different genotypes of Dengue viruses in mice. Sci Rep. 2016;6:19953. doi: 10.1038/srep19953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.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(4):444–51. doi: 10.4269/ajtmh.1989.40.444 [DOI] [PubMed] [Google Scholar]
PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0013550.r001

Decision Letter 0

David Safronetz, Joshua Anzinger

16 Mar 2025

Antibody responses to Japanese encephalitis and dengue viruses in children from a flavivirus endemic region after IMOJEV vaccination

PLOS Neglected Tropical Diseases

Dear Dr. Vista,

Thank you for submitting your manuscript to PLOS Neglected Tropical Diseases. After careful consideration, we feel that it has merit but does not fully meet PLOS Neglected Tropical Diseases's publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript within 60 days May 15 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosntds@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pntd/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

* A rebuttal letter that responds to each point raised by the editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'. This file does not need to include responses to any formatting updates and technical items listed in the 'Journal Requirements' section below.

* A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

* An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, competing interests statement, or data availability statement, please make these updates within the submission form at the time of resubmission. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

We look forward to receiving your revised manuscript.

Kind regards,

Joshua Anzinger

Academic Editor

PLOS Neglected Tropical Diseases

David Safronetz

Section Editor

PLOS Neglected Tropical Diseases

Shaden Kamhawi

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

orcid.org/0000-0003-4304-636XX

Paul Brindley

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

orcid.org/0000-0003-1765-0002

Additional Editor Comments :

While all reviewer comments should be addressed in full, major methodological issues were raised by two reviewers, particularly in relation to assays used to confirm DENV neutralization. There was also concern regarding the lack of assessment of other DENV serotypes. The methodological issues should be addressed with additional experimental work that provides greater strength to the author’s conclusions.

Journal Requirements:

1) We do not publish any copyright or trademark symbols that usually accompany proprietary names, eg ©,  ®, or TM  (e.g. next to drug or reagent names). Therefore please remove all instances of trademark/copyright symbols throughout the text, including:

- ® on page: 8

- TM on pages: 7, and 8.

2) We have noticed that you have uploaded Supporting Information files, but you have not included a list of legends. Please add a full list of legends for your Supporting Information files after the references list.

3) We notice that your supplementary figures are uploaded with the file type 'Figure'. Please amend the file type to 'Supporting Information'. Please ensure that each Supporting Information file has a legend listed in the manuscript after the references list.

4) We note that your Data Availability Statement is currently as follows: "All relevant data are within the manuscript and its Supporting Information files.". Please confirm at this time whether or not your submission contains all raw data required to replicate the results of your study. Authors must share the “minimal data set” for their submission. PLOS defines the minimal data set to consist of the data required to replicate all study findings reported in the article, as well as related metadata and methods (https://journals.plos.org/plosone/s/data-availability#loc-minimal-data-set-definition).

For example, authors should submit the following data: 

1) The values behind the means, standard deviations and other measures reported;

2) The values used to build graphs;

3) The points extracted from images for analysis..

Authors do not need to submit their entire data set if only a portion of the data was used in the reported study.

If your submission does not contain these data, please either upload them as Supporting Information files or deposit them to a stable, public repository and provide us with the relevant URLs, DOIs, or accession numbers. For a list of recommended repositories, please see https://journals.plos.org/plosone/s/recommended-repositories. 

If there are ethical or legal restrictions on sharing a de-identified data set, please explain them in detail (e.g., data contain potentially sensitive information, data are owned by a third-party organization, etc.) and who has imposed them (e.g., an ethics committee). Please also provide contact information for a data access committee, ethics committee, or other institutional body to which data requests may be sent. If data are owned by a third party, please indicate how others may request data access.

5) Please ensure that the funders and grant numbers match between the Financial Disclosure field and the Funding Information tab in your submission form. Note that the funders must be provided in the same order in both places as well. Currently, the order of the grants is different in both places.

Reviewers' Comments:

Reviewer's Responses to Questions

Key Review Criteria Required for Acceptance?

As you describe the new analyses required for acceptance, please consider the following:

Methods

-Are the objectives of the study clearly articulated with a clear testable hypothesis stated?

-Is the study design appropriate to address the stated objectives?

-Is the population clearly described and appropriate for the hypothesis being tested?

-Is the sample size sufficient to ensure adequate power to address the hypothesis being tested?

-Were correct statistical analysis used to support conclusions?

-Are there concerns about ethical or regulatory requirements being met?

Reviewer #1: In this study, the authors intended to investigate (1) if the prior DENV infection would affect the JEV vaccination titer and (2) if the JEV vaccination-induced antibody could enhance DENV infection. The authors concluded that “while JEV vaccination increases DENV-binding antibodies, this cross-reactivity does not lead to virus enhancement”. However, the way that the data being presented caused confusion, such as presenting D2VLP-ELISA results as an indication of prior DENV exposure rather than presenting neutralization results. Furthermore, the authors conclude that “while live JEV vaccination increases DENV-binding antibody, this cross-reactivity does not lead to virus enhancement.” The only data to support such a conclusion is the enhancement of DENV-2 RVP without showing the other serotypes of DENV. The overall sample size is also too small to draw significant conclusions.

Reviewer #2: (No Response)

Reviewer #3: The methodologies described were sound and appropriately referenced.

The study is well designed, and all limitations were discussed.

**********

Results

-Does the analysis presented match the analysis plan?

-Are the results clearly and completely presented?

-Are the figures (Tables, Images) of sufficient quality for clarity?

Reviewer #1: 1. The gold standard of defining the prior DENV infection is by performing neutralizing assay. However, throughout the manuscript, including the title, the authors mentioned DENV antibodies, which were actually measured only by DENV-2 RVP. This is a big concern regarding the study design and the conclusion drawn here. First, although the authors provided the D2-VLP-ELISA results, considering its poor sensitivity, how many infants having DENV exposure pre-vaccination need to be determined using all four serotypes of DENV (RVP preferred to be consistent). Secondly, it would make more sense if figure 2E and 2F presented as DENV neutralization positive or negative using all four serotypes of DENV as the indication of prior DENV infection. Third, in figure 3C-3F, it looks like the same groups of infants with pre-vaccinated DENV2 binding antibody have the neutralizing activity against DENV2 RVP and showed enhancement DENV-2 pre and post vaccination (n=8). Therefore, no statistical significance of D2 enhancement is quite clear. However, no results were shown for other serotypes of DENV. Fourth, since these infants also respond to JEV vaccination with good JEV antibody titers (except B27), the polyclonal sera include both DENV (not limited to DENV2) and JEV antibodies, performing antigen-specific IgG depletion assay by using DENV or JEV from pre- or post-vaccination sera would answer the question if JEV vaccine-induced antibody could enhance the DENV infection and vice versa.

2. Line 253-254, the authors concluded that the lack of sensitivity of JEV-VLP-based ELISA in detecting vaccine-induced antibodies. No reference has been cited or any preliminary data was performed to determine the validity of the assay, such as how much antigens are needed for coating, why 10 or 25 ng/well for JEV or D2. I wonder if the authors tried to use commercial kit for validation.

3. In this study, the authors used RVP with the backbone from WNV but the structure protein gene derived from either DENV-2 (strain 16681) or JEV (Strain 7812474). Since strain 16681 is an old strain and no longer circulates, the authors should test the enhancement of current circulation strains in the Philippines to reflect the concern of vaccine-induced severe diseases. Similarly, please provide further information if strain 7812474 is similar to the JEV circulating in the Philippines and if it belongs to the same genotype of vaccine strain SA14-14-2.

4. Did the authors acquire information about whether the recruited subjects received Dengvaxia or YFV 17D vaccination before? Such information should be revealed in the manuscript.

5. The statistical tests require further consultation with the statistician. For example, why use a one-tailed student t-test or Spearman test instead of a two-tailed one? On several occasions, when the sample size is small, the Fisher exact test should be explored.

6. Could the authors explain why only 17 subjects were selected for post vaccination DENV antibody reactivity experiments since the total 29 subjects were not too big?

7. The discussion on Line 360-367 tends to over-explain the data.

Reviewer #2: (No Response)

Reviewer #3: the results are well presented, clear and cohesive with the analysis plan

**********

Conclusions

-Are the conclusions supported by the data presented?

-Are the limitations of analysis clearly described?

-Do the authors discuss how these data can be helpful to advance our understanding of the topic under study?

-Is public health relevance addressed?

Reviewer #1: (No Response)

Reviewer #2: (No Response)

Reviewer #3: The findings are described well without overstating the conclusion.

**********

Editorial and Data Presentation Modifications?

Use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity. If the only modifications needed are minor and/or editorial, you may wish to recommend “Minor Revision” or “Accept”.

Reviewer #1: 1. Throughout the manuscript, flaviruses should be corrected as orthoflaviviruses based on the recent ICTV classification

2. Line 70-71, not just DENV and JEV are prevalent in tropical countries. ZIKV as well and should be included in the sentence.

3. Line 86, DENV is “carried” should be corrected as “transmitted”

Reviewer #2: (No Response)

Reviewer #3: Nil

**********

Summary and General Comments

Use this section to provide overall comments, discuss strengths/weaknesses of the study, novelty, significance, general execution and scholarship. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. If requesting major revision, please articulate the new experiments that are needed.

Reviewer #1: (No Response)

Reviewer #2: The manuscript written by Vista et al. describes about analyzing antibody response patterns against DENV and JEV after immunization with attenuated-chimeric JEV vaccine. The serum samples were collected from Filipino children and were subjected to ELISA, neutralization test and ADE assay. Their results suggested that IMOJEV did not induce ADE against DENV-2 in DENV-seronegative children and did not affect the pattern of dose-response enhancing activity curves in the seropositive individuals. Authors concluded that IMOJEV were able to induce neutralizing antibody against JEV in Filipino children without induction of ADE against DENV-2. Although the sample size is small as mentioned by authors, the present information may be important for children and their parents who are concerning and doubting whether JEV vaccine might induce ADE or not. Specific comments and suggestions are described below.

In this paper, antibody levels were mostly indicated by ELISA ratio and PRNT50 titer, and both data were mixed across figures, which sometimes made it difficult to grasp the whole story. For instance, in Fig. 1C, JEV antibody levels were determined by ELISA for pre-vaccination samples, while in Fig. 2C, JEV antibody levels were determined by PRNT for post-vaccination samples. This way may not bring benefits to readers for comparing data between pre- and post-vaccination samples. Such expression needs to be improved.

I did not feel very much the necessity of ELISA data in this paper, since NT activity was demonstrated to be strongly correlated with ADE activity. To persuade the need for ELISA data, please show correlation in antibody level between ELISA ratio and PRNT50 titer when DENV antigen was used (as well JEV antigen was used).

Minor comment

Statement about Fig. 3F is missing from the manuscript.

Reviewer #3: This manuscript is very well written with good study aims and significance.

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #2: No

Reviewer #3: Yes:  Chuan Kok Lim

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

Figure resubmission:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step. If there are other versions of figure files still present in your submission file inventory at resubmission, please replace them with the PACE-processed versions.

Reproducibility:

?>

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0013550.r003

Decision Letter 1

David Safronetz, Joshua Anzinger

11 Aug 2025

Response to Reviewers Revised Manuscript with Track Changes Manuscript

Shaden Kamhawi

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

orcid.org/0000-0003-4304-636XX

Paul Brindley

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

orcid.org/0000-0003-1765-0002

Additional Editor Comments: Reviewers' comments:

Key Review Criteria Required for Acceptance?

As you describe the new analyses required for acceptance, please consider the following:

Methods:

-Are the objectives of the study clearly articulated with a clear testable hypothesis stated?

-Is the study design appropriate to address the stated objectives?

-Is the population clearly described and appropriate for the hypothesis being tested?

-Is the sample size sufficient to ensure adequate power to address the hypothesis being tested?

-Were correct statistical analysis used to support conclusions?

-Are there concerns about ethical or regulatory requirements being met?

Reviewer #2: (No Response)

Reviewer #3: (No Response)

Reviewer #4: Please see my comments

Results:

-Does the analysis presented match the analysis plan?

-Are the results clearly and completely presented?

-Are the figures (Tables, Images) of sufficient quality for clarity?

Reviewer #2: (No Response)

Reviewer #3: (No Response)

Reviewer #4: Please see my comments

Conclusions:

-Are the conclusions supported by the data presented?

-Are the limitations of analysis clearly described?

-Do the authors discuss how these data can be helpful to advance our understanding of the topic under study?

-Is public health relevance addressed?

Reviewer #2: (No Response)

Reviewer #3: (No Response)

Reviewer #4: Please see my comments

Editorial and Data Presentation Modifications?

Use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity. If the only modifications needed are minor and/or editorial, you may wish to recommend “Minor Revision” or “Accept”.

Reviewer #2: (No Response)

Reviewer #3: (No Response)

Reviewer #4: Please see my comments

Summary and General Comments:

Use this section to provide overall comments, discuss strengths/weaknesses of the study, novelty, significance, general execution and scholarship. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. If requesting major revision, please articulate the new experiments that are needed.

Reviewer #2: (No Response)

Reviewer #3: The authors have appropriately addressed the reviewers' comments

Reviewer #4: This is an interesting study to assess the Ab responses to a live JEV vaccine administered to young Philippino children (9-24 months of age) living in an area with endemic circulation of DENVs, JEV, Zika. The investigators conducted the study to support broader use of JEV vaccines in the country and to address concerns about a JEV vaccine enhancing dengue.

The main questions they address consist of

1) Does pre-existing flavivirus immunity suppress or enhance the JEV vaccine response?

2) Does the JEV vaccine stimulate cross reactive binding and functional Abs to dengue type 2?

3) Does the JEV vaccine stimulate antibodies with the potential to enhance DV2 infections

The investigators obtain clear answers to the first 2 questions. They observe that prior flavivirus immunity has no impact on vaccine take or magnitude of the JEV NAb response. They also observe no boosting of DV2 NAbs by the JEV vaccine.

The studies to answer the 3rd question are misguided and poorly interpreted. Cell culture ADE assays have no predictive value for immune enhanced dengue disease. See my comments below.

Finally, while this is a straightforward study, the results are poorly presented, and the reader is left quite confused about the results and their implications. See my comments below.

Specific comments

1) The investigators appear to be unfamiliar with basic aspects of flavivirus serology, and the study is not presented in a proper context. The investigators determine dengue immune status before vaccination by testing for DV2 binding Abs. They observe 8/28 DV2 positive children. For the entire paper they consider these children to be DV2 immune. If they are consistent, they would also call these children JEV immune because 7/8 are also positive for JEV binding Abs. The VLP binding assays do not have the required specificity for determining virus-specific immunity because of extensive cross reactivity. Please use the term flavivirus immune because the binding Abs could be the result of a past exposure to any DV serotype, JEV or Zika. The assay for determining virus specific immunity is the neutralization assay. Only 3/8 children with binding Abs actually neutralized DV2. Please revise the paper to reflect the fact that 8/28 kids were flavi immune at baseline and only 3 of these kids were actually DV2 immune.

2) The finding that nearly all the children respond to the vaccine and develop JEV NAb, with minimal impact on DV2 NAb is important but lost in the way the results are presented. Please consider reorganizing the figures by presenting results relevant to specific questions? In the current paper, many graphs with different comparisons are presented in an indiscriminate manner.

3) The JEV vaccine drives a NAb response in nearly all children, but this is not reflected in the JEV VLP binding assay, where many children remain negative. This could be due to vaccine induced IgM competing with IgG or a technical issue with the assay. Please discuss.

4) As already mentioned, cell culture ADE is not predictive of risk in people. Even the cell culture ADE measurements are confusing. The investigators choose fold enhancement. Why do some sera enhance at the highest concentration tested while others require significant dilution of the serum. If one serum sample enhanced 5 fold at a dilution of 1:20 only and another serum reaches a peak of 5 fold enhancement at a serum dilution of 1:200, as presente din this paper they both have a peak fold enhancement of 5. Are they really the same? If the authors look closely at the ADE curves, they will see that kids with high DV2 neut titers have to be diluted to enhance while those with no NAb sometimes enhance at the highest concertation tested. What does this mean with respect to JEV vaccination enhancing DV2 in vivo? The only message I take from these data are that the cell culture ADE profiles remain the same before and after vaccination suggesting they are being driven by immunity that existed before vaccination and not Abs stimulated by the vaccine.

I hope these comments are useful to reorganize and rewrite this paper to highlight the important results and conclusions of this study.

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #2: No

Reviewer #3: Yes:  Dr Chuan Kok Lim

Reviewer #4: No

Figure resubmission:Reproducibility:--> -->-->To enhance the reproducibility of your results, we recommend that authors of applicable studies deposit laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols-->?>

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0013550.r005

Decision Letter 2

David Safronetz, Joshua Anzinger

9 Sep 2025

Dear Dr. Vista,

We are pleased to inform you that your manuscript 'Antibody responses to Japanese encephalitis virus and dengue virus serotype 2 in children from an orthoflavivirus endemic region after IMOJEV vaccination' has been provisionally accepted for publication in PLOS Neglected Tropical Diseases.

Before your manuscript can be formally accepted you will need to complete some formatting changes, which you will receive in a follow up email. A member of our team will be in touch with a set of requests.

Please note that your manuscript will not be scheduled for publication until you have made the required changes, so a swift response is appreciated.

IMPORTANT: The editorial review process is now complete. PLOS will only permit corrections to spelling, formatting or significant scientific errors from this point onwards. Requests for major changes, or any which affect the scientific understanding of your work, will cause delays to the publication date of your manuscript.

Should you, your institution's press office or the journal office choose to press release your paper, you will automatically be opted out of early publication. We ask that you notify us now if you or your institution is planning to press release the article. All press must be co-ordinated with PLOS.

Thank you again for supporting Open Access publishing; we are looking forward to publishing your work in PLOS Neglected Tropical Diseases.

Best regards,

Joshua Anzinger

Academic Editor

PLOS Neglected Tropical Diseases

David Safronetz

Section Editor

PLOS Neglected Tropical Diseases

Shaden Kamhawi

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

orcid.org/0000-0003-4304-636XX

Paul Brindley

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

orcid.org/0000-0003-1765-0002

***********************************************************

p.p1 {margin: 0.0px 0.0px 0.0px 0.0px; line-height: 16.0px; font: 14.0px Arial; color: #323333; -webkit-text-stroke: #323333}span.s1 {font-kerning: none

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0013550.r006

Acceptance letter

David Safronetz, Joshua Anzinger

Dear Dr. Vista,

We are delighted to inform you that your manuscript, "Antibody responses to Japanese encephalitis virus and dengue virus serotype 2 in children from an orthoflavivirus endemic region after IMOJEV vaccination," has been formally accepted for publication in PLOS Neglected Tropical Diseases.

We have now passed your article onto the PLOS Production Department who will complete the rest of the publication process. All authors will receive a confirmation email upon publication.

The corresponding author will soon be receiving a typeset proof for review, to ensure errors have not been introduced during production. Please review the PDF proof of your manuscript carefully, as this is the last chance to correct any scientific or type-setting errors. Please note that major changes, or those which affect the scientific understanding of the work, will likely cause delays to the publication date of your manuscript. Note: Proofs for Front Matter articles (Editorial, Viewpoint, Symposium, Review, etc...) are generated on a different schedule and may not be made available as quickly.

Soon after your final files are uploaded, the early version of your manuscript will be published online unless you opted out of this process. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

You will receive an invoice from PLOS for your publication fee after your manuscript has reached the completed accept phase. If you receive an email requesting payment before acceptance or for any other service, this may be a phishing scheme. Learn how to identify phishing emails and protect your accounts at https://explore.plos.org/phishing.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Neglected Tropical Diseases.

Best regards,

Shaden Kamhawi

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

Paul Brindley

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Pre- and post- IMOJEV vaccination anti-JEV neutralizing titers.

    (TIF)

    pntd.0013550.s001.tif (969.4KB, tif)
    S2 Fig. Relationship between the log10 JEV RVPNT50 titers and (A) age or (B) DENV2 binding reactivities (two-tailed Spearman rank correlation test).

    (TIF)

    pntd.0013550.s002.tif (76.1KB, tif)
    S3 Fig. Correlation between DENV2 ELISA P/N Values and DENV 1, 3 and 4 ELISA P/N Values.

    (TIF)

    pntd.0013550.s003.tif (467.7KB, tif)
    S4 Fig. Pre- and post-IMOJEV vaccination anti-DENV2 neutralizing titers.

    (TIF)

    pntd.0013550.s004.tif (471.9KB, tif)
    S5 Fig. Pre- and post-IMOJEV vaccination DENV2 antibody dependent enhancement of selected samples.

    (TIF)

    pntd.0013550.s005.tif (314.9KB, tif)
    S6 Fig. Comparison of vaccine response based on pre-IMOJEV vaccination DENV2 functional antibody response.

    (TIF)

    pntd.0013550.s006.tif (39.7KB, tif)
    S7 Fig. Correlation between (A) DENV2 or (B) JEV ELISA P/N values with neutralization titers.

    (TIF)

    pntd.0013550.s007.tif (623.7KB, tif)
    S1 Table. Master table of ELISA, neutralization and enhancement assay results.

    (XLSX)

    pntd.0013550.s008.xlsx (12.6KB, xlsx)
    S2 Table. Test of different JEV antigen concentrations for selected serum samples at 1:200 serum dilution.

    (XLSX)

    pntd.0013550.s009.xlsx (9.4KB, xlsx)
    Attachment

    Submitted filename: Response to Reviewers.docx

    pntd.0013550.s011.docx (41.7KB, docx)
    Attachment

    Submitted filename: 2025_08_011Response to Reviewers+ALR.docx

    pntd.0013550.s012.docx (28.6KB, docx)

    Data Availability Statement

    Data Availability Statement: All relevant data are within the manuscript and its Supporting information files, with the exception of exact participant age, which is potentially identifying. Access to this data is restricted by the University of the Philippines Manila Research Ethics Board (UPMREB) to protect participant confidentiality. Data may be made available upon reasonable request to the UPMREB (upmreb@post.upm.edu.ph).


    Articles from PLOS Neglected Tropical Diseases are provided here courtesy of PLOS

    RESOURCES