Advertisement
Research Article| Volume 129, P142-151, April 2023

Antibody cross-reactivity and evidence of susceptibility to emerging Flaviviruses in the dengue-endemic Brazilian Amazon

Open AccessPublished:January 31, 2023DOI:https://doi.org/10.1016/j.ijid.2023.01.033

      Highlights

      • Over 75% of adult individuals had antidengue antibodies in the city of Manaus.
      • Dengue virus-positive individuals do not cross-neutralize zika virus efficiently.
      • Varying degree of cross-reactivity against emerging and endemic Flaviviruses.

      Abstract

      Objectives

      Several Flaviviruses can co-circulate. Pre-existing immunity to one virus can modulate the response to a heterologous virus; however, the serological cross-reaction between these emerging viruses in dengue virus (DENV)-endemic regions are poorly understood.

      Methods

      A cross-sectional study was performed among the residents of Manaus city in the state of Amazonas, Brazil. The serological response was assessed by hemagglutination inhibition assay (HIA), enzyme-linked immunosorbent assay, and neutralization assay.

      Results

      A total of 74.52% of the participants were immunoglobulin G-positive (310/416), as estimated by lateral flow tests. Overall, 93.7% of the participants were seropositive (419/447) for at least one DENV serotype, and the DENV seropositivity ranged between 84.8% and 91.0%, as determined by HIA. About 93% had antiyellow fever virus 17D-reactive antibodies, whereas 80.5% reacted to wild-type yellow fever virus. Zika virus (ZIKV) had the lowest seropositivity percentage (52.6%) compared with other Flaviviruses. Individuals who were DENV-positive with high antibody titers by HIA or envelope protein domain III enzyme-linked immunosorbent assay reacted strongly with ZIKV, whereas individuals with low anti-DENV antibody titers reacted poorly toward ZIKV. Live virus neutralization assay with ZIKV confirmed that dengue serogroup and ZIKV-spondweni serogroup are far apart; hence, individuals who are DENV-positive do not cross-neutralize ZIKV efficiently.

      Conclusion

      Taken together, we observed a high prevalence of DENV in the Manaus-Amazon region and a varying degree of cross-reactivity against emerging and endemic Flaviviruses. Epidemiological and exposure conditions in Manaus make its population susceptible to emerging and endemic arboviruses.

      Keywords

      Introduction

      Emerging arboviruses are a growing health problem in Brazil and worldwide. During the last decade, Brazil has endured the zika virus (ZIKV), chikungunya virus, and yellow fever virus (YFV) epidemics. These pathogens have not only been associated with morbidity and mortality but also with additional health care cost. Arboviruses, such as dengue virus (DENV) and YFV, have been endemic in Brazil, whereas YFV is endemic/enzootic for centuries being introduced during the slave traffic. DENV was introduced recently and has been endemic for the last 50 years. However, the recent YFV epidemic in Southeast Brazil demands a better understanding on the transmission dynamics and disease distribution in general population [
      • Vasconcelos PFdC
      Yellow fever in Brazil: thoughts and hypotheses on the emergence in previously free areas.
      ].
      The introduction of ZIKV in Brazil caused a huge epidemic in some regions of Brazil, which was associated with microcephaly in the newborns [
      • Brasil P
      • et al.
      Zika virus infection in pregnant women in Rio de Janeiro.
      ,
      • Mlakar J
      • et al.
      Zika virus associated with microcephaly.
      ] and Guillain-Barré syndrome and a few fatal cases in adults [
      • dos Santos T
      • et al.
      Zika virus and the Guillain–Barré syndrome — case series from seven countries.
      ,
      • de Oliveira WK
      • et al.
      Zika virus infection and associated neurologic disorders in Brazil.
      ]. The high density of Aedes mosquitoes in several cities was responsible for the high transmission rates; however, little is understood about the interaction between DENV and ZIKV and the underlying cause of the magnitude of the ZIKV epidemic in Brazil.
      Previous DENV infection increases the risk of future dengue with warning signs and severe dengue. Non-neutralizing antibodies against the envelope (E) protein induced during the primary DENV infection can cross-react with other DENV serotypes or heterologous Flaviviruses. These DENV cross-reactive antibodies can facilitate subsequent secondary or tertiary heterologous DENV infection of myeloid cells due the antibody-dependent enhancement (ADE) and can increase dengue disease severity in humans [
      • Goncalvez AP
      • et al.
      Monoclonal antibody-mediated enhancement of dengue virus infection in vitro and in vivo and strategies for prevention.
      ]. Similarly, DENV serotypes and other Flaviviruses share peptides that can modulate cluster of differentiation (CD; CD4+ and CD8+) T cell responses upon an infection with a heterologous Flavivirus [
      • Grifoni A
      • et al.
      Prior dengue virus exposure shapes T cell immunity to zika virus in humans.
      ].
      In vitro and mouse challenge studies have shown that antibodies raised against DENV can enhance ZIKV infection [
      • Bardina SV
      • et al.
      Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity.
      ,
      • Zimmerman MG
      • et al.
      Cross-reactive dengue virus antibodies augment zika virus infection of human placental macrophages.
      ]. However, previous DENV infection was not associated with ZIKV viremia or cytokine expression in experimentally challenged macaques [
      • McCracken MK
      • et al.
      Impact of prior Flavivirus immunity on Zika virus infection in rhesus macaques.
      ,
      • Pantoja P
      • et al.
      Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus.
      ,
      • Breitbach ME
      • et al.
      Primary infection with dengue or Zika virus does not affect the severity of heterologous secondary infection in macaques.
      ] or in humans [
      • Terzian ACB
      • et al.
      Viral load and cytokine response profile does not support antibody-dependent enhancement in dengue-primed zika virus-infected patients.
      ,
      • Santiago GA
      • et al.
      Prior dengue virus infection is associated with increased viral load in patients infected with dengue but not zika virus.
      ,
      • Michlmayr D
      • et al.
      Comprehensive immunoprofiling of pediatric zika reveals key role for monocytes in the acute phase and no effect of prior dengue virus infection.
      ] nor with fetal demise or congenital Zika syndrome in pregnant women [
      • Halai UA
      • et al.
      Maternal zika virus disease severity, virus load, prior dengue antibodies, and their relationship to birth outcomes.
      ,
      • Damasceno L
      • et al.
      Why did ZIKV perinatal outcomes differ in distinct regions of Brazil? An exploratory study of two cohorts.
      ]. Emerging evidence suggests that previous DENV infection may not enhance noncongenital Zika disease, but whether previous ZIKV infection increases future dengue disease in humans remains unknown. In this study, we evaluated the antibody cross-reactivity against Flaviviruses in a dengue-endemic population to understand the role of pre-existing antibodies in modulating secondary heterologous Flavivirus infection. Here, we also estimated the seropositivity rates against 10 different endemic and emerging Flaviviruses in Manaus, capital of the Amazonas state.

      Methods

      Study population and sample and data collection

      The study population comprised healthy individuals accessing services at the Centro de Controle de Zoonoses, Manaus-Amazonas. A total of 450 consecutive participants were recruited between January 2015 and December 2015. The study included individuals of both sexes aged ≥18 years who agreed to participate. Study participants signed the written informed consent form and answered an epidemiological questionnaire. A total of 4 ml of venous blood was drawn from each participant using ethylenediaminetetraacetic acid tubes (BD Vacutainer). Soon after blood collection, the collection tubes were centrifuged, and plasma was separated and stored at -80°C until further analysis.

      Sample size calculation

      A sample size of 398 individuals was calculated using an estimated prevalence of 50% and 95% confidence interval for a large population with a desired precision of 0.01 [
      • Salgado BB
      • et al.
      Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
      ]. To achieve an adequate sampling without significant dropouts due to incomplete questionnaires and subsequent reduction in statistical power, we recruited 450 individuals. However, three questionnaires were incomplete and removed from the analysis. Sample size calculation was performed using the Epi Info software version 7.2.

      Maps and socioeconomic indicators from Brazil

      Maps were created using the QGIS Software version 2.18.26 for macOS. Graphs displaying the Human Development Indexes and Sanitation Indicators were extracted from public database, Atlas Brasil from the 2010 Census (https://atlasbrasil.org.br) and the National System of Sanitation Information/Trata Brasil from 2018 (http://www.snis.gov.br), respectively.

      Dengue rapid test

      Lateral flow point of care (POC) DENV antibodies detection test was performed using the BioPix® Dengue IgM/IgG Rapid test (Wama Diagnóstica, São Paulo, Brazil) at the Laboratory of Emerging Viruses at the University of Campinas (LEVE - Unicamp). The plasma samples were assayed to qualitatively detect immunoglobulin (Ig)G and IgM antibodies against all four DENV serotypes. The assay had a sensitivity of 99% and specificity of 98% as declared by the manufacturer. The detection of IgG in this assay was developed to detect low levels of IgG.

      Hemagglutination inhibition test

      To assess the overall Flavivirus reactivity, hemagglutination inhibition assay (HIA) was performed at the Evandro Chagas Institute, Belém. The plasma samples collected were subjected to an HIA and adapted to the microplate technique, with a titration cut-off point of 1/20, as previously described [
      • Salgado BB
      • et al.
      Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
      ]. The plasma samples were tested for antibodies against the YFV strain (YFV-17D) and 10 endemic or emerging arboviruses of the Flavivirus genus in the Brazilian Amazon: YFV, DENV serotypes 1-4 (DENV-1, DENV-2, DENV-3, and DENV-4), ZIKV, Saint Louis Encephalitis virus (SLEV), West Nile virus (WNV), Ilheus virus (ILHV), and Rocio virus (ROCV).

      Cloning, expression, and purification of recombinant DENV2 and ZIKV envelope domain III proteins

      The DENV2 envelope domain III protein sequence (E-DIII, residues 577-674, GenBank number HQ026763) was cloned and expressed using the pET28b vector (Silvia Beatriz Boscardin, University of São Paulo), as previously described [
      • Amaral MP
      • et al.
      Homologous prime-boost with Zika virus envelope protein and poly (I:C) induces robust specific humoral and cellular immune responses.
      ]. The ZIKV envelope amino acid sequences from different isolates were obtained from GenBank, and multiple sequence alignments were performed to identify an amino acid consensus sequence using MEGA software version 7.0.26. A polyhistidine (6x-His) tag was added at the N-terminal to facilitate downstream purification. Consensus ZIKV E-DIII sequence was codon-optimized and inserted into prokaryotic expression vector pGS21-a (GenOne Biotechnologies, Brazil).

      Enzyme-linked immunosorbent assay (ELISA) to detect anti-DENV and ZIKV E-DIII-specific IgG antibodies

      We standardized an indirect in-house ELISA to detect anti-DENV and anti-ZIKV IgG antibodies in plasma samples using recombinant E-DIII proteins as antigens, as previously described [
      • Lalwani P
      • et al.
      SARS-CoV-2 seroprevalence and associated factors in Manaus, Brazil: baseline results from the DETECTCoV-19 cohort study.
      ].

      ZIKV neutralization test

      Focus reduction neutralization test (FRNT) was performed in collaboration with the Laboratory of Emerging Viruses at University of Campinas, as previously described [
      • Silva-Filho JL
      • et al.
      Gas6 drives Zika virus-induced neurological complications in humans and congenital syndrome in immunocompetent mice.
      ]. In brief, the plasma samples were heated at 56°C for 30 minutes to inactivate the complement system. Four-fold plasma samples diluted with Earle minimum essential medium (Sigma-Aldrich) were incubated with 100 focus forming units of ZIKV (strain BeH823339-Asian). Virus antigen was identified by an indirect immunoassay, and focus forming units were counted for each plasma dilution and the results were tabulated.

      Statistical analysis

      Descriptive statistics was used to describe the sociodemographic features and mosquito preventive measures of the study population. The missing values were excluded from the calculations. Bubble charts showing the percentages of seropositives with HIA antibody titers were created with Microsoft Excel 2019 software. The 95% confidence intervals were computed using the Blaker method. All statistical analyses were performed using the GraphPad Prism software version 9.1.2. A paired t-test was used to evaluate the ELISA results for DENV- and ZIKV-E-DIII. The linear correlation between variables was determined using Pearson correlation. P-values ≤0.05 were considered statistically significant.

      Results

      High dengue prevalence in urban Manaus

      A random convenience sampling strategy enrolled 450 individuals in Manaus; this noninterventional cross-sectional study between January and December 2015 (Supplementary Figure 1). A total of 447 healthy individuals of both sexes and aged ≥18 years were included in the final analysis; three questionnaires were incomplete and removed from the analysis. The demographic features of the study population are described in Table 1. We oversampled females (70.5%) and the median age of our study population was 38 years. A total of 30% of the study participants self-declared that they were unemployed in the last 12 months and 61.70% had completed high school (Table 1). A total of 37.5% and 13.2% of the study participants self-reported a previous dengue and malaria infection, respectively. A total of 80.8% reported vaccination for YFV and 75.9% use SUS-Brazil's public health system. A majority (94.9%) reported performing at least one preventive measure against mosquitoes: avoiding standing water and properly disposing of trash, followed by personal mosquito repellents, which were the most popular measures adapted to prevent mosquitoes (Table 2).
      Table 1Sociodemographic features of study population- Manaus, Amazonas.
      CharacteristicValue %
      Age (years)18-197.80
      20-2923.10
      30-3924.70
      40-4923.60
      ≥5020.80
      SexFemale70.50
      Family size13.50
      214.80
      324.20
      425.60
      ≥531.90
      Number of children in house052.90
      129.00
      211.30
      33.90
      ≥42.90
      Occupation
      Last 12 months
      Unemployed31.40
      Domestic service workers and manual workers14.00
      Professionals with university education10.60
      Professionals or technicians10.10
      Administrative service workers10.10
      Poorly specified occupations of informal work9.60
      Service and commercial workers8.70
      Member of the armed forces, police and military firefighters2.50
      Senior government officials or senior corporate officials1.60
      Art Professional1.40
      Family IncomeMinimum wage22.60
      Two minimum wages29.80
      Three minimum wages20.50
      Four minimum wages or more27.10
      Education levelIlliterate1.30
      Literate7.40
      Elementary school9.80
      High school61.70
      Graduate19.80
      a Last 12 months
      Table 2Mosquito prevention practices reported by study participants.
      CharacteristicValue %
      Dengue infection
      Self-reported
      Yes37.5
      Malaria infection
      Self-reported
      Yes13.2
      Vaccinated against Yellow fever
      Self-reported
      Yes80.8
      Use Sistema Único de Saúde (SUS)
      Brazilian public funded health care system
      Yes75.9
      Neighbors with dengue feverYes43.3
      Mosquito problem in neighborhoodYes43.1
      Perform preventive measures against mosquitoesYes94.9
      Mosquito prevention measures
      Multiple responses
      :
      Avoid standing water64.30
      Disposing trash correctly62.50
      Personal mosquito repellents: cream and spray54.90
      Personal and household hygiene33.70
      Fans28.10
      Close windows and doors21.80
      Mosquito coil, mats and liquid vaporizers12.60
      Mosquito net for door and window10.90
      Protective clothing9.40
      Mosquito bed nets5.80
      Others4.30
      a Self-reported
      b Brazilian public funded health care system
      c Multiple responses
      Manaus is a tropical metropolis in Brazil with about 2.5 million inhabitants in the middle of the Amazon rainforest (Figure 1a). Manaus, the capital of the Amazonas state, compared with other Brazilian capitals, shows a low sociodemographic and human development index. In Manaus, the lack of sanitation is also widespread, with wastewater facilitating vector proliferation and waterborne diseases, which affect the poorest residents (Figure 1b). Figure 1c compares our study population age distribution compared with the 2010 census data.
      Figure 1
      Figure 1Elevated dengue seropositivity rates in urban Manaus.
      (a) Maps depict location of Manaus. (b) HDI (left) and Sanitation indicators (right) for the 27 Brazilian capitals were obtained from the 2010 census and a 2018 survey, respectively. (c) The study population distribution is compared to 2010 Census data obtained from the Brazilian Institute of Geography and Statistics (IBGE). (d-e) Plasma samples were tested for anti-DENV antibodies using BioPix® Dengue Rapid Test IgG/IgM (n = 416) and HIA (n = 447) and seropositivity percentages with 95% confidence interval were plotted. BioPix® Dengue Rapid Test IgG/IgM was qualitative. HIA was quantitative and samples with HIA titer ≥20 units were considered positive for the test virus.
      DENV, dengue virus; HDI, Human Development Index; HIA, hemagglutination inhibition assay.
      Next, we assessed the prevalence of anti-DENV antibodies using lateral flow POC rapid tests and HIA. A total of 74.52% of participants were IgG-positive (310/416) and 1.68% (15/416) had anti-DENV IgM antibodies, as estimated by POC tests. Overall, 93.7% of participants were seropositive (419/447) for at least one DENV serotype and the DENV seropositivity ranged between 84.8% and 91.0%, as determined by HIA (Figure 1d and Table 3). We observed that most of our participants aged over 20 years had anti-DENV antibodies (Figure 1e and Supplementary Figure 2).
      Table 3Prevalence of anti-Flavivirus antibodies.
      SerogroupVirus
      Flaviviridae (Flavivirus genus): YFV, DENV serotypes 1 to 4 (DENV-1, DENV-2, DENV-3 and DENV-4), ZIKV, WNV, SLEV, ILHV, ROCV. DENV, dengue virus; ILHV, ilheus virus; ROCV, rocio virus; SLEV, Saint Louis Encephalitis virus; WNV, West Nile virus; YFV, yellow fever virus; ZIKV, zika virus.
      Total
      Hemagglutination Inhibition titer ≥1:20
      %
      Age (years)
      18-1920-2930-3940-49≥50
      Yellow fever vaccineYFV/17D92.8 (415/447)67.6 (23/34)98.0 (99/101)95.4 (103/108)93.2 (96/103)92.3 (84/91)
      YFV GroupYFV80.5 (360/447)61.8 (21/34)77.2 (78/101)83.3 (90/108)79.6 (82/103)86.8 (79/91)
      DENV GroupDENV
      Positive for at least one DENV serotype
      93.7 (419/447)79.4 (27/34)95.0 (96/101)95.4 (103/108)94.2 (97/103)95.6 (87/91)
      DENV-190.6 (405/447)76.5 (26/34)93.1 (94/101)91.7 (99/108)88.3 (91/103)93.4 (85/91)
      DENV-291.0 (407/447)82.3 (28/34)93.1 (94/101)92.6 (100/108)88.3 (91/103)92.3 (84/91)
      DENV-385.5 (382/447)82.3 (28/34)86.1 (87/101)87.0 (94/108)84.5 (87/103)83.5 (76/91)
      DENV-484.8 (379/447)64.7 (22/34)87.1 (88/101)88.0 (95/108)84.5 (87/103)84.6 (77/91)
      Spondweni GroupZIKV52.6 (235/447)32.3 (11/34)48.5 (49/101)55.5 (60/108)54.4 (56/103)59.3 (54/91)
      Japanese Encephalitis Virus GroupWNV81.9 (366/447)55.9 (19/34)84.2 (85/101)88.0 (95/108)78.6 (81/103)84.6 (77/91)
      SLEV86.3 (386/447)64.7 (22/34)87.1 (88/101)89.8 (97/108)84.5 (87/103)90.1 (82/91)
      Ntaya Virus GroupILHV85.7 (383/447)64.7 (22/34)85.1 (86/101)90.7 (98/108)83.5 (86/103)89.0 (81/91)
      ROCV75.8 (339/447)70.6 (24/34)74.3 (75/101)72.2 (78/108)79.6 (82/103)78.0 (71/91)
      Negatives3.4 (15/447)11.8 (4/34)4.0 (4/101)0.9 (1/108)3.9 (4/103)2.2 (2/91)
      a Hemagglutination Inhibition titer ≥1:20
      b Positive for at least one DENV serotype
      c Flaviviridae (Flavivirus genus): YFV, DENV serotypes 1 to 4 (DENV-1, DENV-2, DENV-3 and DENV-4), ZIKV, WNV, SLEV, ILHV, ROCV.DENV, dengue virus; ILHV, ilheus virus; ROCV, rocio virus; SLEV, Saint Louis Encephalitis virus; WNV, West Nile virus; YFV, yellow fever virus; ZIKV, zika virus.

      Cross-reactivity between principal endemic and emerging Flaviviruses

      We performed HIA, a cell-based assay, to assess the overall cross-reactivity by performing serial dilution of the plasma samples; the results are summarized in Figure 2 and Table 3. YFV is endemic in the Amazon region and vaccination is compulsory. Here, we observed that 92.8% of individuals tested have anti-YFV-17D reactive antibodies; although, only 80.3% self-reported receiving YFV vaccine (Table 2) compared with 80.5% toward the wild-type YFV (Table 3). A total of 37.2% self-reported previous DENV infection, whereas we detected 93.7% of individuals with anti-DENV antibodies against at least one DENV serotype. A chi-square test found a significant difference between the observed (laboratory-confirmed) and expected (self-reported) dengue infection (P <0.0001, χ2 test, data not shown). Among the DENV serotypes, DENV-2, followed by DENV-1, DENV-3, and DENV-4 had the highest seropositivity (Figure 2 and Table 3). Individuals with high DENV titers correspondingly had higher reactivity to other Flaviviruses (Figure 2a). All samples were collected before or during the ZIKV epidemic in Manaus, and we observed that ZIKV, among all viruses tested, had the lowest seropositivity percentage (Figure 2b). The percentage of individuals reactive toward Flaviviruses classified into serogroups is presented in Table 3. Overall, antibody reactivity was the lowest among the 18-19 years age group and increased rapidly with age for all arboviruses tested (Table 3, Figure 2, and Supplementary Figure 2).
      Figure 2
      Figure 2Antibody reactivity profile against emerging and re-emerging human Flaviviruses.
      Samples (n = 447) were tested for antibodies against 10 different endemic and emerging Flaviviruses and yellow fever vaccine strain using the HIA. (a) Colors depict the HIA titers, each column represents one patient, and each row is one test virus on the heat map. Samples described in the heatmap were sorted as per DENV-2 HIA titers. (b) Samples with HIA titer ≥20 units were considered positive and plotted as percentage positives among the study age groups. (c) HIA titers were compared to participant age. Solid lines are splines for each test virus.
      DENV, dengue virus; HIA, hemagglutination inhibition assay; ILHV, ilheus virus; ROCV, rocio virus; SLEV, Saint Louis Encephalitis virus; WNV, West Nile virus; YFV, yellow fever virus; ZIKV, zika virus.

      Low cross-reactivity between preexisting anti-DENV antibodies and ZIKV

      Next, we evaluated the cross-reactivity to ZIKV among individuals who were DENV-positive using HIA, ELISA, and live virus neutralization assay. First, modified bubble plots compared DENV-1, DENV-2, or ZIKV HIA titers and their distribution for each virus to the reference virus. Upon comparing the HIA titers, we observed that most of the individuals who were DENV-positive with low titers failed to react with ZIKV. Whereas only individuals with elevated titers for DENV-1 or DENV-2 reacted with ZIKV in the HIA assay (Figure 3).
      Figure 3
      Figure 3Low cross-reactivity between pre-existing antidengue antibodies and ZIKV.
      Modified bubble charts compare cross-reactivity (n = 447), (a) DENV-1 with DENV-2 or ZIKV, and (b) DENV-2 with DENV-1 or ZIKV. All samples with HIA titer ≥20 units were considered positive. Bubble size is proportional to the percentage of individuals with the corresponding HIA titer, and each column adds up to 100%. HIA titers are color coded for the first reference virus column for comparison. Each pie chart represents the composition of HIA titer for the reference test virus.
      DENV, dengue virus; HIA, hemagglutination inhibition assay; ZIKV, zika virus.
      E-DIII is an important target for neutralizing antibodies among Flaviviruses; hence, we performed an ELISA using E-DIII proteins for DENV-2 or ZIKV and assessed antibody reactivities. Similarly, in HIA, we observed that anti-DENV2 E-DIII IgG levels were elevated compared with anti-ZIKV E-DIII IgG antibodies (Figure 4a). A paired analysis of the samples showed a significant decrease in the antibody reactivities between DENV and ZIKV (Figure 4b-c). As expected, anti-DENV E-DIII antibody levels estimated by ELISA significantly increased with age (Figure 4d); and stratified by sex, males showed the highest reactivity toward DENV-2 envelope protein domain III (Figure 4e). Splines depict the relationship between HIA antibody titers and ELISA reactivity (Figure 4f).
      Figure 4
      Figure 4Reactivity between DENV and ZIKV envelope domain III proteins.
      ELISA plates were coated with DENV-2 or ZIKV envelope domain III (E-DIII) proteins to detect immunoglobulin G antibodies in human plasma samples (n = 416). (a) Range of antibody reactivity toward E-DIII antigens is depicted. The red horizontal lines denote median OD (450 nm) values. (b) Pairwise comparison denotes the reactivity toward DENV-2 and ZIKV. Mann Whitney or paired t-test, **** P <0.0001. (c) DENV-2 and ZIKV anti-E-DIII antibody levels measured by ELISA were compared, red line in the graph represents the spline fitting for the distribution. (d) DENV-2 or ZIKV E-DIII reactivity was compared to participant (d) age or (e) sex. Solid lines represent splines fit for the antibody distribution. Pearson's correlation and P-values are denoted in the graphs. (f) HIA tiers for dengue virus serotypes are compared between DENV-2 or ZIKV E-DIII reactivity determined by ELISA. Each solid line is color coded and represents one test virus. Each dot represents one sample in all the graphs.
      DENV, dengue virus; E-DIII, envelope protein domain III; ELISA, enzyme-linked immunosorbent assay; HIA, hemagglutination inhibition assay; OD450, optical density, 450 nm; ZIKV, zika virus.
      Furthermore, to understand the DENV antibody reactivity and its relationship to a possible neutralizing antibody response to ZIKV, we performed an FRNT with live ZIKV in a subset (n = 57) of DENV-positive samples chosen at random. Figure 5a compares DENV-2 or ZIKV ED-III ELISA results with ZIKV FRNT50 values; samples were divided as higher or lower than the median of reactivity toward the DENV-2 ED-III antigen. Primarily, we observed that individuals with anti-DENV E-DIII OD >median (red) neutralized ZIKV more efficiently than individuals with anti-DENV E-DIII OD <median (green). The percentage relative infection of ZIKV was used to calculate neutralizing antibody titers; each patient is represented by one curve (Figure 5b-d). Overall, the DENV-2 E-DIII antibody levels are inversely proportional to the ZIKV FRNT values, and individuals with high DENV reactive antibodies had a potential neutralizing activity against ZIKV. Collectively, these results indicate that both the quantity and quality of DENV-specific responses are necessary to neutralize ZIKV.
      Figure 5
      Figure 5High titers of DENV reactive antibodies neutralize ZIKV.
      Focus reduction neutralization test (FRNT) for ZIKV was performed to estimate neutralizing antibodies in patient plasma samples (n = 57). (a) DENV-2 or ZIKV E-DIII protein specific immunoglobulin G reactivities are depicted in first two columns. Box and whiskers indicate the median lines, 25 and 75- interquartile and min-to-max points. Red dots denote samples with anti-DENV-2 E-DIII reactivity values greater than the median value and green dots denote samples displaying anti-DENV-2 reactivity less than the median value. Third column in the graph represents the 50-percent FRNT (FRNT50) values for ZIKV. Right y-axis is FRNT50 values in log for ZIKV. ZIKV antibody neutralization dose-response curves were plotted with (b) high (OD450 > median) or (c) low (OD450 < median) DENV-2 E-DIII antibody reactivity. Each curve represents one patient sample diluted serially. Anti-ZIKV NS1 monoclonal antibody was used as control. (d) Representative neutralization dose-response curves were plotted based on DENV-2 E-DIII antibody reactivity.
      DENV, dengue virus; E-DIII, envelope protein domain III; FRNT, focus reduction neutralization test; OD450, optical density, 450 nm; ZIKV, zika virus.

      Discussion

      Endemic and emerging arboviruses are a huge public health concern worldwide not only due to their unexpected clinical manifestations and potential complications but also due to the lack of appropriate diagnostic assays and vaccines [
      • Mota MTdO
      • et al.
      Mosquito-transmitted viruses - the great Brazilian challenge.
      ,
      • Musso D
      • Desprès P.
      Serological diagnosis of Flavivirus-associated human infections.
      ,
      • Pierson TC
      • Diamond MS.
      The continued threat of emerging flaviviruses.
      ]. In this study, we report one of the highest dengue seropositivity in Manaus-Brazil [
      • Salgado BB
      • et al.
      Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
      ]; 74.52% by POC and 93.7% by HIA. This confirms that most residents of the region have likely been infected with DENV, if not with other arboviruses at least once. Evidence of varying degree of antibody cross-reactivity with principal emerging Flaviviruses depends on the virus serogroup and pre-existing level of antibody titers. Thus, individuals with the highest anti-DENV antibody titers could cross-neutralize ZIKV more efficiently than individuals with low anti-DENV titers. Elevated dengue prevalence in Manaus and the Amazon region does not undermine the threat from other endemic and emerging arboviruses.
      Dengue is endemic in Brazil for at least 50 years, and without adequate prevention and treatment, the number of reported cases of dengue has increased in Brazil and other countries in the Americas [
      • Salles TS
      • et al.
      History, epidemiology and diagnostics of dengue in the American and Brazilian contexts: a review.
      ,
      • Nunes PCG
      • et al.
      30 years of fatal dengue cases in Brazil: a review.
      ]. In our previous study, we observed an increase in seroprevalence in several Brazilian cities in the last decades [
      • Salgado BB
      • et al.
      Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
      ]. On the contrary, dengue incidence levels have been the same over the last decade in most regions; nevertheless, there has been a huge increase in severe dengue disease cases and deaths [
      • Salles TS
      • et al.
      History, epidemiology and diagnostics of dengue in the American and Brazilian contexts: a review.
      ,
      • Nunes PCG
      • et al.
      30 years of fatal dengue cases in Brazil: a review.
      ,
      • Teixeira MG
      • et al.
      Epidemiological trends of dengue disease in Brazil (2000–2010): a systematic literature search and analysis.
      ,
      • Fares RC
      • et al.
      Epidemiological scenario of dengue in Brazil.
      ]. Three of four Brazilian municipalities are heavily infested with the mosquito Aedes aegypti, which highlights the staggering epidemiological and economic burden in the endemic regions [
      • Mota MTdO
      • et al.
      Mosquito-transmitted viruses - the great Brazilian challenge.
      ,
      • Tapia-Conyer R
      • et al.
      Dengue: an escalating public health problem in Latin America.
      ]. Thus, the prospects of controlling dengue or other arbovirus diseases are not promising. All four DENV serotypes have been detected in Manaus, with the late introduction of DENV-3 and DENV-4 [
      • Salgado BB
      • et al.
      Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
      ,
      • de Souza Bastos M
      • et al.
      Simultaneous circulation of all four dengue serotypes in Manaus, State of Amazonas, Brazil in 2011.
      ,
      • Nava A
      • et al.
      The impact of global environmental changes on infectious disease emergence with a focus on risks for Brazil.
      ]. Our serological results demonstrate a lower seropositivity toward DENV-3 and DENV-4 than DENV-2 and DENV-1. The high DENV prevalence in urban Manaus observed in this study is in line with our previous observations from other Brazilian cities [
      • Salgado BB
      • et al.
      Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
      ]. A total of 37.2% of individuals self-reported a previous DENV infection compared with over 75% individuals with detectable antibodies in this study. This disparity between the observed and expected numbers could be largely related to asymptomatic or mild cases that have gone unnoticed and highlights the need for more laboratory diagnosis of febrile cases.
      Yellow fever vaccination is compulsory in Manaus and in the Amazon region, and 92.8% of the study participants had anti-YFV-17D reactive antibodies, whereas 80.5% had anti-YFV reactive antibodies against the wild-type virus. ILHV and ROCV have been endemic in Brazil; however, their spread in the Amazon region and prevalence studies are lacking. In this current study, the Ntaya virus group, represented by ILHV and ROCV, had a seropositivity rate of 85.7% and 75.8%, respectively. Recently, WNV [
      • Martins LC
      • et al.
      First isolation of West Nile virus in Brazil.
      ,
      • Costa ÉA
      • Giovanetti M
      • Silva Catenacci L
      • et al.
      West nile virus in Brazil.
      ] and SLEV [
      • Weaver SC
      • et al.
      Isolation of Saint Louis encephalitis virus from a horse with neurological disease in Brazil.
      ,
      • Silva JR
      • et al.
      A Saint Louis encephalitis and Rocio virus serosurvey in Brazilian horses.
      ] cases have been reported among horses in Brazil, but no human cases have been reported. The co-circulation of these arboviruses that share B and T cell epitopes provides critical insight into the role of pre-existing immunity essential to assess the clinical implications of the disease. Infection with DENV can protect against the onset of apparent signs and severe illness after subsequent JEV, SLEV, and WNV infections [
      • Sather GE
      • Hammon WM.
      Protection against St. Louis encephalitis and West Nile arboviruses by previous dengue virus (types 1–4) infection.
      ,
      • Price WH
      • Thind IS.
      Protection against West Nile virus induced by a previous injection with dengue virus.
      ,
      • Tarr GC
      • Hammon WM.
      Cross-protection between group B arboviruses: resistance in mice to Japanese B encephalitis and St. Louis encephalitis viruses induced by dengue virus immunization.
      ]; ZIKV infection also confers protection against subsequent WNV infection and associated symptoms [
      • Vázquez-Calvo Á
      • et al.
      Zika virus infection confers protection against West Nile virus challenge in mice.
      ]; JEV and SLEV infections can protect against severe disease and mortality after WNV and DENV infections [
      • Goverdhan MK
      • et al.
      Two-way cross-protection between West Nile and Japanese encephalitis viruses in bonnet macaques.
      ,
      • Tesh RB
      • et al.
      Immunization with heterologous flaviviruses protective against fatal West Nile encephalitis.
      ,
      • Petrovsky N
      • et al.
      An inactivated cell culture Japanese encephalitis vaccine (JE-ADVAX) formulated with delta inulin adjuvant provides robust heterologous protection against West Nile encephalitis via cross-protective memory B cells and neutralizing antibody.
      ,
      • Li J
      • et al.
      Cross-protection induced by Japanese encephalitis vaccines against different genotypes of Dengue viruses in mice.
      ]; and ILHV and SLEV can elicit cross-protection against a lethal ROCV challenge [
      • Amarilla AA
      • et al.
      Ilheus and Saint Louis encephalitis viruses elicit cross-protection against a lethal Rocio virus challenge in mice.
      ]. Compulsory YFV vaccination and the high prevalence of DENV in Manaus may partially explain the lack of evidence for co-circulation of other Flaviviruses. This cross-protection may partly also explain why YFV is not present in India and other Asian countries, which are endemic to WNV, JEV, DENV, and other Flaviviruses [
      • Monath TP
      • Vasconcelos PFC.
      Yellow fever.
      ]. On the other hand, the recent ZIKV outbreak in India demonstrates that cross-protection might depend on the residual concentration of cross-reactive antibodies, suggesting that individual antibody titer set point rather than waning could indicate disease risk [
      • Katzelnick LC
      • et al.
      Dengue and Zika virus infections in children elicit cross-reactive protective and enhancing antibodies that persist long term.
      ,
      • Gurav YK
      • et al.
      First case of Zika virus infection during an outbreak of chikungunya in a rural region of Maharashtra state, India.
      ].
      The samples used in this study were collected before or during the ZIKV epidemic in Brazil. In this current study, 52.6% had anti-ZIKV antibodies, the lowest seropositivity rate among all the viruses tested by HIA. Also, E-DIII ELISA demonstrated a weak reactivity toward ZIKV among individuals who were DENV-positive. Thus, a substantial proportion of participants showed evidence of exposure to DENV but not ZIKV [
      • Langerak T
      • et al.
      The possible role of cross-reactive dengue virus antibodies in Zika virus pathogenesis.
      ]. Moreover, individuals who were DENV-positive with low HIA titers failed to react with ZIKV and only high-titer DENV plasma samples reacted with ZIKV. The role of previous DENV infection in ADE has been controversial; nevertheless, there is little evidence of severe disease after ZIKV infection in individuals who were DENV-positive [
      • Bardina SV
      • et al.
      Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity.
      ,
      • Zimmerman MG
      • et al.
      Cross-reactive dengue virus antibodies augment zika virus infection of human placental macrophages.
      ,
      • McCracken MK
      • et al.
      Impact of prior Flavivirus immunity on Zika virus infection in rhesus macaques.
      ,
      • Pantoja P
      • et al.
      Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus.
      ,
      • Breitbach ME
      • et al.
      Primary infection with dengue or Zika virus does not affect the severity of heterologous secondary infection in macaques.
      ,
      • Terzian ACB
      • et al.
      Viral load and cytokine response profile does not support antibody-dependent enhancement in dengue-primed zika virus-infected patients.
      ,
      • Santiago GA
      • et al.
      Prior dengue virus infection is associated with increased viral load in patients infected with dengue but not zika virus.
      ,
      • Michlmayr D
      • et al.
      Comprehensive immunoprofiling of pediatric zika reveals key role for monocytes in the acute phase and no effect of prior dengue virus infection.
      ,
      • Halai UA
      • et al.
      Maternal zika virus disease severity, virus load, prior dengue antibodies, and their relationship to birth outcomes.
      ,
      • Damasceno L
      • et al.
      Why did ZIKV perinatal outcomes differ in distinct regions of Brazil? An exploratory study of two cohorts.
      ]. The patterns of antibody cross-neutralization suggest that ZIKV lies outside the DENV serocomplex [
      • Montoya M
      • et al.
      Longitudinal analysis of antibody cross-neutralization following zika virus and dengue virus infection in Asia and the Americas.
      ]; similarly, we observed that HIA was able to distinguish ZIKV from DENV infections when all viruses were analyzed simultaneously. We observed a lack of durable cross-neutralizing antibody response against ZIKV from individuals who were DENV-positive, as previously reported [
      • Collins MH
      • et al.
      Lack of durable cross-neutralizing antibodies against zika virus from dengue virus infection.
      ]. In this study, patients with the highest anti-DENV antibody titers neutralized ZIKV more efficiently, which is in line with a previous report demonstrating that individuals with elevated DENV titers due to repeated exposure to dengue virus elicit robust cross-neutralizing antibodies against ZIKV [
      • Hattakam S
      • et al.
      Repeated exposure to dengue virus elicits robust cross neutralizing antibodies against Zika virus in residents of Northeastern Thailand.
      ].
      In Manaus, a high prevalence of DENV serotypes and compulsory YFV vaccination together may provide a strong increase in homologous and heterologous cross-neutralizing antibodies. Zika vaccination in an individual who had DENV has been shown to boost pre-existing Flavivirus immunity and elicit protective responses against both ZIKV and DENV [
      • Dussupt V
      • et al.
      Potent Zika and dengue cross-neutralizing antibodies induced by Zika vaccination in a dengue-experienced donor.
      ]. On the other hand, tick-borne encephalitis virus vaccination in YFV vaccinated individuals caused a significant reduction in the tick-borne encephalitis-specific neutralizing antibodies [
      • Bradt V
      • et al.
      Pre-existing yellow fever immunity impairs and modulates the antibody response to tick-borne encephalitis vaccination.
      ]. T cell responses seem to be robust between Flaviviruses and may, at least in part, explain the cross-protection seen against ZIKV from DENV infection [
      • Subramaniam KS
      • et al.
      Two is better than one: evidence for T-cell cross-protection between dengue and zika and implications on vaccine design.
      ]. Nevertheless, the durability of humoral and cellular response needs to be assessed in longitudinal studies to determine the role of the pre-existing immunity in protection or pathology.
      Due to many positive samples, we were unable to perform a statistical analysis to identify epidemiological risk factors. Our convenience sampling oversampled females which impede calculation of true dengue prevalence for Manaus city; however, our results demonstrate that urban Manaus DENV prevalence is high; most of the individuals get infected before attaining age 36 years and the male sex had higher antibody titers. We used DENV2 ED-III protein in ELISA to compare with ZIKV ED-III response and did not test with other dengue serotypes; nevertheless, we believe that a similar relationship will be observed because DENV and ZIKV belong to distinct serogroups. Although DENV-neutralizing serotype-specific antibodies are mainly against E-DIII, there might be a sustainable antibody response against other targets, such as domains I/II of the envelope protein, which were not evaluated in this study. We also did not have history of hospitalization or severe disease caused by dengue among our study participants to correlate with the humoral response. A longitudinal study in Nicaragua comparing DENV and ZIKV infection observed that primary infection increases the cross-reactive antibodies, which decline slowly over time; however, postsecondary infection the decay rate is much slower [
      • Katzelnick LC
      • et al.
      Dengue and Zika virus infections in children elicit cross-reactive protective and enhancing antibodies that persist long term.
      ]. In addition, secondary infection with a different DENV serotype or ZIKV infection can alter the antibody kinetics and levels [
      • Montoya M
      • et al.
      Longitudinal analysis of antibody cross-neutralization following zika virus and dengue virus infection in Asia and the Americas.
      ]. In this cross-sectional study, we were able to map the cross-reactivity for emerging and endemic Flaviviruses. However, a longitudinal study would be necessary to understand the persistence of antibody titers after viral infection and the role of reinfection to maintain or increase immune response to the exposed pathogens.
      In conclusion, we observed a low cross-protection among individuals who were DENV-positive to ZIKV. However, further longitudinal studies are needed on ADE and the possible role of cross-reactive anti-DENV antibodies in Flavivirus pathogenesis. ZIKV and other emerging Flaviviruses will become increasingly prevalent in DENV-endemic regions, raising the possibility that pre-existing immunity to one virus could modulate the response to a heterologous virus; although, whether this would be beneficial or detrimental remains unclear and could vary with the combination of the viruses prevalent in the region. The COVID-19 pandemic has negatively affected mosquito control measures; in addition, financial resources have been diverted toward the pandemic, making control measures more difficult. Currently, Brazil and the Amazonian region face a complex epidemiological scenario characterized by simultaneous circulation of several arboviruses and high mosquito density; hence, a functional vector control program and febrile syndrome surveillance are essential to identify unforeseen epidemiological threats.

      CRediT authorship contribution statement

      Barbara Batista Salgado: Conceptualization, Methodology, Formal analysis, Investigation, Data curation, Writing – review & editing, Visualization. Fábio Carmona de Jesus Maués: Conceptualization, Methodology, Formal analysis, Investigation, Writing – original draft. Maele Jordão: Methodology, Investigation. Renato Lemos Pereira: Methodology, Formal analysis, Investigation. Daniel A. Toledo-Teixeira: Methodology, Formal analysis, Investigation. Pierina L. Parise: Methodology, Investigation. Fabiana Granja: Methodology, Investigation. Higo Fernando Santos Souza: Resources. Marcio Massao Yamamoto: Resources. Jannifer Oliveira Chiang: Methodology, Investigation. Livia Caricio Martins: Resources. Silvia Beatriz Boscardin: Resources. Jaila Dias Borges Lalwani: Writing – review & editing, Supervision. Pedro Fernando C Vasconcelos: Resources, Funding acquisition. José Luiz Proença-Modena: Conceptualization, Formal analysis, Resources, Writing – review & editing, Funding acquisition. Pritesh Lalwani: Conceptualization, Formal analysis, Writing – review & editing, Project administration, Funding acquisition.

      Declaration of competing interest

      The authors have no competing interests to declare.

      Funding

      Pritesh Lalwani was supported by grants from Fundação de Amparo à Pesquisa do Estado do Amazonas (FAPEAM), PPSUS, and CT&I ÁREAS PRIORITÁRIAS research program. Pedro Fernando C Vasconcelos is fellow of the National Council for Scientific and Technological Development (CNPq) and supported by grants. José Luiz Proença Modena was financially supported by the São Paulo Research Foundation (FAPESP) 2016/00194-8 and 2020/04558-0, CNPq 305628/2020-8, and Fundo de Apoio ao Ensino, Pesquisa e Extensão (FAEPEX) 2266/20 and 0002/20. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of this manuscript. The authors thank FAPEAM and Programa de Apoio à Pós-Graduação Stricto Sensu (POSGRAD) for their support. Bárbara Batista Salgado and Fábio Carmona de Jesus Maués received a scholarship from Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Pierina L Parise received a scholarship from CAPES and FAPESP, and Daniel A. Toledo Teixeira received a scholarship from CNPq.

      Ethical approval

      This observational and cross-sectional study was approved by the research ethics committee of the Universidade Federal do Amazonas, with approval number CAAE 96171218.7.0000.5020, in accordance with the Brazilian law, which complied with the Declaration of Helsinki. All study participants gave oral and written informed consent before enrollment.

      Appendix. Supplementary materials

      References

        • Vasconcelos PFdC
        Yellow fever in Brazil: thoughts and hypotheses on the emergence in previously free areas.
        Rev Saude Publica. 2010; 44: 1144-1149https://doi.org/10.1590/s0034-89102010005000046
        • Brasil P
        • et al.
        Zika virus infection in pregnant women in Rio de Janeiro.
        N Engl J Med. 2016; 375: 2321-2334https://doi.org/10.1056/NEJMoa1602412
        • Mlakar J
        • et al.
        Zika virus associated with microcephaly.
        N Engl J Med. 2016; 374: 951-958https://doi.org/10.1056/NEJMoa1600651
        • dos Santos T
        • et al.
        Zika virus and the Guillain–Barré syndrome — case series from seven countries.
        N Engl J Med. 2016; 375: 1598-1601https://doi.org/10.1056/NEJMc1609015
        • de Oliveira WK
        • et al.
        Zika virus infection and associated neurologic disorders in Brazil.
        N Engl J Med. 2017; 376: 1591-1593https://doi.org/10.1056/NEJMc1608612
        • Goncalvez AP
        • et al.
        Monoclonal antibody-mediated enhancement of dengue virus infection in vitro and in vivo and strategies for prevention.
        Proc Natl Acad Sci U S A. 2007; 104: 9422-9427https://doi.org/10.1073/pnas.0703498104
        • Grifoni A
        • et al.
        Prior dengue virus exposure shapes T cell immunity to zika virus in humans.
        J Virol. 2017; 91: e01417-e01469https://doi.org/10.1128/JVI.01469-17
        • Bardina SV
        • et al.
        Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity.
        Science. 2017; 356: 175-180https://doi.org/10.1126/science.aal4365
        • Zimmerman MG
        • et al.
        Cross-reactive dengue virus antibodies augment zika virus infection of human placental macrophages.
        Cell Host Microbe. 2018; 24 (e6): 731-742https://doi.org/10.1016/j.chom.2018.10.008
        • McCracken MK
        • et al.
        Impact of prior Flavivirus immunity on Zika virus infection in rhesus macaques.
        PLoS Pathog. 2017; 13e1006487https://doi.org/10.1371/journal.ppat.1006487
        • Pantoja P
        • et al.
        Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus.
        Nat Commun. 2017; 8: 15674https://doi.org/10.1038/ncomms15674
        • Breitbach ME
        • et al.
        Primary infection with dengue or Zika virus does not affect the severity of heterologous secondary infection in macaques.
        PLoS Pathog. 2019; 15e1007766https://doi.org/10.1371/journal.ppat.1007766
        • Terzian ACB
        • et al.
        Viral load and cytokine response profile does not support antibody-dependent enhancement in dengue-primed zika virus-infected patients.
        Clin Infect Dis. 2017; 65: 1260-1265https://doi.org/10.1093/cid/cix558
        • Santiago GA
        • et al.
        Prior dengue virus infection is associated with increased viral load in patients infected with dengue but not zika virus.
        Open Forum Infect Dis. 2019; 6https://doi.org/10.1093/ofid/ofz320
        • Michlmayr D
        • et al.
        Comprehensive immunoprofiling of pediatric zika reveals key role for monocytes in the acute phase and no effect of prior dengue virus infection.
        Cell Rep. 2020; 31107569https://doi.org/10.1016/j.celrep.2020.107569
        • Halai UA
        • et al.
        Maternal zika virus disease severity, virus load, prior dengue antibodies, and their relationship to birth outcomes.
        Clin Infect Dis. 2017; 65: 877-883https://doi.org/10.1093/cid/cix472
        • Damasceno L
        • et al.
        Why did ZIKV perinatal outcomes differ in distinct regions of Brazil? An exploratory study of two cohorts.
        Viruses. 2021; 13https://doi.org/10.3390/v13050736
        • Salgado BB
        • et al.
        Prevalence of arbovirus antibodies in young healthy adult population in Brazil.
        Parasit Vectors. 2021; 14: 403https://doi.org/10.1186/s13071-021-04901-4
        • Amaral MP
        • et al.
        Homologous prime-boost with Zika virus envelope protein and poly (I:C) induces robust specific humoral and cellular immune responses.
        Vaccine. 2020; 38: 3653-3664https://doi.org/10.1016/j.vaccine.2020.03.037
        • Lalwani P
        • et al.
        SARS-CoV-2 seroprevalence and associated factors in Manaus, Brazil: baseline results from the DETECTCoV-19 cohort study.
        Int J Infect Dis. 2021; 110: 141-150https://doi.org/10.1016/j.ijid.2021.07.017
        • Silva-Filho JL
        • et al.
        Gas6 drives Zika virus-induced neurological complications in humans and congenital syndrome in immunocompetent mice.
        Brain Behav Immun. 2021; 97: 260-274https://doi.org/10.1016/j.bbi.2021.08.008
        • Mota MTdO
        • et al.
        Mosquito-transmitted viruses - the great Brazilian challenge.
        Braz J Microbiol. 2016; 47: 38-50https://doi.org/10.1016/j.bjm.2016.10.008
        • Musso D
        • Desprès P.
        Serological diagnosis of Flavivirus-associated human infections.
        Diagnostics (Basel). 2020; 10: 302https://doi.org/10.3390/diagnostics10050302
        • Pierson TC
        • Diamond MS.
        The continued threat of emerging flaviviruses.
        Nat Microbiol. 2020; 5: 796-812https://doi.org/10.1038/s41564-020-0714-0
        • Salles TS
        • et al.
        History, epidemiology and diagnostics of dengue in the American and Brazilian contexts: a review.
        Parasit Vectors. 2018; 11: 264https://doi.org/10.1186/s13071-018-2830-8
        • Nunes PCG
        • et al.
        30 years of fatal dengue cases in Brazil: a review.
        BMC Public Health. 2019; 19: 329https://doi.org/10.1186/s12889-019-6641-4
        • Teixeira MG
        • et al.
        Epidemiological trends of dengue disease in Brazil (2000–2010): a systematic literature search and analysis.
        PLoS Negl Trop Dis. 2013; 7: e2520https://doi.org/10.1371/journal.pntd.0002520
        • Fares RC
        • et al.
        Epidemiological scenario of dengue in Brazil.
        BioMed Res Int. 2015; 2015321873https://doi.org/10.1155/2015/321873
        • Tapia-Conyer R
        • et al.
        Dengue: an escalating public health problem in Latin America.
        Paediatr Int Child Health. 2012; 32: 14-17https://doi.org/10.1179/2046904712Z.00000000046
        • de Souza Bastos M
        • et al.
        Simultaneous circulation of all four dengue serotypes in Manaus, State of Amazonas, Brazil in 2011.
        Rev Soc Bras Med Trop. 2012; 45: 393-394https://doi.org/10.1590/s0037-86822012000300022
        • Nava A
        • et al.
        The impact of global environmental changes on infectious disease emergence with a focus on risks for Brazil.
        ILAR J. 2017; 58: 393-400https://doi.org/10.1093/ilar/ilx034
        • Martins LC
        • et al.
        First isolation of West Nile virus in Brazil.
        Mem Inst Oswaldo Cruz. 2019; 114e180332https://doi.org/10.1590/0074-02760180332
        • Costa ÉA
        • Giovanetti M
        • Silva Catenacci L
        • et al.
        West nile virus in Brazil.
        Pathogens. 2021; 10: 896https://doi.org/10.3390/pathogens10070896
        • Weaver SC
        • et al.
        Isolation of Saint Louis encephalitis virus from a horse with neurological disease in Brazil.
        PLOS Negl Trop Dis. 2013; 7: e2537https://doi.org/10.1371/journal.pntd.0002537
        • Silva JR
        • et al.
        A Saint Louis encephalitis and Rocio virus serosurvey in Brazilian horses.
        Rev Soc Bras Med Trop. 2014; 47: 414-417https://doi.org/10.1590/0037-8682-0117-2014
        • Sather GE
        • Hammon WM.
        Protection against St. Louis encephalitis and West Nile arboviruses by previous dengue virus (types 1–4) infection.
        Proc Soc Exp Biol Med. 1970; 135: 573-578https://doi.org/10.3181/00379727-135-35098a
        • Price WH
        • Thind IS.
        Protection against West Nile virus induced by a previous injection with dengue virus.
        Am J Epidemiol. 1971; 94: 596-607https://doi.org/10.1093/oxfordjournals.aje.a121358
        • Tarr GC
        • Hammon WM.
        Cross-protection between group B arboviruses: resistance in mice to Japanese B encephalitis and St. Louis encephalitis viruses induced by dengue virus immunization.
        Infect Immun. 1974; 9: 909-915https://doi.org/10.1128/iai.9.5.909-915.1974
        • Vázquez-Calvo Á
        • et al.
        Zika virus infection confers protection against West Nile virus challenge in mice.
        Emerg Microbes Infect. 2017; 6: e81https://doi.org/10.1038/emi.2017.68
        • Goverdhan MK
        • et al.
        Two-way cross-protection between West Nile and Japanese encephalitis viruses in bonnet macaques.
        Acta Virol. 1992; 36: 277-283
        • Tesh RB
        • et al.
        Immunization with heterologous flaviviruses protective against fatal West Nile encephalitis.
        Emerg Infect Dis. 2002; 8: 245-251https://doi.org/10.3201/eid0803.010238
        • Petrovsky N
        • et al.
        An inactivated cell culture Japanese encephalitis vaccine (JE-ADVAX) formulated with delta inulin adjuvant provides robust heterologous protection against West Nile encephalitis via cross-protective memory B cells and neutralizing antibody.
        J Virol. 2013; 87: 10324-10333https://doi.org/10.1128/JVI.00480-13
        • Li J
        • et al.
        Cross-protection induced by Japanese encephalitis vaccines against different genotypes of Dengue viruses in mice.
        Sci Rep. 2016; 6: 19953https://doi.org/10.1038/srep19953
        • Amarilla AA
        • et al.
        Ilheus and Saint Louis encephalitis viruses elicit cross-protection against a lethal Rocio virus challenge in mice.
        PLoS One. 2018; 13e0199071https://doi.org/10.1371/journal.pone.0199071
        • Monath TP
        • Vasconcelos PFC.
        Yellow fever.
        J Clin Virol. 2015; 64: 160-173https://doi.org/10.1016/j.jcv.2014.08.030
        • Katzelnick LC
        • et al.
        Dengue and Zika virus infections in children elicit cross-reactive protective and enhancing antibodies that persist long term.
        Sci Transl Med. 2021; 13: eabg9478https://doi.org/10.1126/scitranslmed.abg9478
        • Gurav YK
        • et al.
        First case of Zika virus infection during an outbreak of chikungunya in a rural region of Maharashtra state, India.
        Trans R Soc Trop Med Hyg. 2022; 116: 974-977https://doi.org/10.1093/trstmh/trac022
        • Langerak T
        • et al.
        The possible role of cross-reactive dengue virus antibodies in Zika virus pathogenesis.
        PLoS Pathog. 2019; 15e1007640https://doi.org/10.1371/journal.ppat.1007640
        • Montoya M
        • et al.
        Longitudinal analysis of antibody cross-neutralization following zika virus and dengue virus infection in Asia and the Americas.
        J Infect Dis. 2018; 218: 536-545https://doi.org/10.1093/infdis/jiy164
        • Collins MH
        • et al.
        Lack of durable cross-neutralizing antibodies against zika virus from dengue virus infection.
        Emerg Infect Dis. 2017; 23: 773-781https://doi.org/10.3201/eid2305.161630
        • Hattakam S
        • et al.
        Repeated exposure to dengue virus elicits robust cross neutralizing antibodies against Zika virus in residents of Northeastern Thailand.
        Sci Rep. 2021; 11: 9634https://doi.org/10.1038/s41598-021-88933-x
        • Dussupt V
        • et al.
        Potent Zika and dengue cross-neutralizing antibodies induced by Zika vaccination in a dengue-experienced donor.
        Nat Med. 2020; 26: 228-235https://doi.org/10.1038/s41591-019-0746-2
        • Bradt V
        • et al.
        Pre-existing yellow fever immunity impairs and modulates the antibody response to tick-borne encephalitis vaccination.
        npj Vaccines. 2019; 4: 38https://doi.org/10.1038/s41541-019-0133-5
        • Subramaniam KS
        • et al.
        Two is better than one: evidence for T-cell cross-protection between dengue and zika and implications on vaccine design.
        Front Immunol. 2020; 11: 517https://doi.org/10.3389/fimmu.2020.00517