Immunogenicity of a Live-Attenuated Dengue Vaccine Using a ...

11
Immunogenicity of a Live-Attenuated Dengue Vaccine Using a Heterologous Prime-Boost Strategy in a Phase 1 Randomized Clinical Trial Leyi Lin, Michael A Koren, Kristopher M Paolino, Kenneth H Eckels, Rafael De La Barrera, Heather Friberg, Jeffrey R Currier, Gregory D Gromowski, Naomi E Aronson, Paul B Keiser, Marvin J Sklar, Erica L Sondergaard, Louis E Jasper, Timothy P Endy, Richard G Jarman, Stephen J Thomas Downloaded from https://academic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 May 2022

Transcript of Immunogenicity of a Live-Attenuated Dengue Vaccine Using a ...

Immunogenicity of a Live-Attenuated Dengue Vaccine

Using a Heterologous Prime-Boost Strategy in a

Phase 1 Randomized Clinical Trial

Leyi Lin, Michael A Koren, Kristopher M Paolino, Kenneth H Eckels, Rafael De La

Barrera, Heather Friberg, Jeffrey R Currier, Gregory D Gromowski, Naomi E

Aronson, Paul B Keiser, Marvin J Sklar, Erica L Sondergaard, Louis E Jasper,

Timothy P Endy, Richard G Jarman, Stephen J Thomas

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

M A J O R A R T I C L E

Dengue Prime-Boost Vaccines • jid 2021:223 (15 May) • 1707

The Journal of Infectious Diseases

Received 23 June 2020; editorial decision 16 September 2020; accepted 18 September 2020; published online September 23, 2020.

Correspondence: Michael Koren, MD, Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910 ([email protected]).

The Journal of Infectious Diseases® 2021;223:1707–16Published by Oxford University Press for the Infectious Diseases Society of America 2020. This work is written by (a) US Government employee(s) and is in the public domain in the US.DOI: 10.1093/infdis/jiaa603

Immunogenicity of a Live-Attenuated Dengue Vaccine Using a Heterologous Prime-Boost Strategy in a Phase 1 Randomized Clinical TrialLeyi Lin,1 Michael A. Koren,1 Kristopher M. Paolino,2 Kenneth H. Eckels,3 Rafael De La Barrera,3 Heather Friberg,1 Jeffrey R. Currier,1 Gregory D. Gromowski,1 Naomi E. Aronson,4 Paul B. Keiser,1 Marvin J. Sklar,5 Erica L. Sondergaard,2 Louis E. Jasper,6 Timothy P. Endy,7 Richard G. Jarman,1 and Stephen J. Thomas1

1Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA, 2Clinical Trials Center, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA, 3Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA, 4Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA, 5Naval Medical Research Center, Silver Spring, Maryland, USA, 6US Army Medical Materiel Development Activity, Frederick, Maryland, USA, and 7SUNY Upstate Medical University, Institute for Global Health and Translational Sciences, Syracuse, New York, USA

Background. Dengue is a global health problem and the development of a tetravalent dengue vaccine with durable protection is a high priority. A heterologous prime-boost strategy has the advantage of eliciting immune responses through different mechanisms and therefore may be superior to homologous prime-boost strategies for generating durable tetravalent immunity. 

Methods. In this phase 1 first-in-human heterologous prime-boost study, 80 volunteers were assigned to 4 groups and received a tetravalent dengue virus (DENV-1–4) purified inactivated vaccine (TDENV-PIV) with alum adjuvant and a tetravalent dengue virus (DENV-1–4) live attenuated vaccine (TDENV-LAV) in different orders and dosing schedules (28 or 180 days apart). 

Results. All vaccination regimens had acceptable safety profiles and there were no vaccine-related serious adverse events. TDEN-PIV followed by TDEN-LAV induced higher neutralizing antibody titers and a higher rate of tetravalent seroconversions compared to TDEN-LAV followed by TDEN-PIV. Both TDEN-PIV followed by TDEN-LAV groups demonstrated 100% tetravalent serocon-version 28 days following the booster dose, which was maintained for most of these subjects through the day 180 measurement. 

Conclusions. A heterologous prime-boost vaccination strategy for dengue merits additional evaluation for safety, immunoge-nicity, and potential for clinical benefit.

Clinical Trials Registration. NCT02239614.Keywords. dengue virus; dengue vaccine; immunogenicity.

Dengue remains the world’s leading mosquito borne illness with an estimated 390 million infections occurring annually, of which over 95 million result in clinically apparent disease [1]. Having an effective tetravalent dengue vaccine is a high priority and the dengue vaccine field is robust with numerous candi-dates in preclinical and clinical development [2]. Several excel-lent reviews on the status of dengue vaccine development have been published [3, 4]. Dengvaxia, a tetravalent dengue vaccine with a yellow fever virus backbone, is the only vaccine that has been licensed and has been registered in over 20 countries. It is a case study on the current difficulties in developing dengue vaccines due to the lack of an optimal animal model that reli-ably translates to human efficacy and the lack of a biomarker that indicates protection [5]. For Dengvaxia, a safety signal

observed in the youngest recipients and those who were dengue seronegative at baseline prompted a change in the original in-dication. Currently, the vaccine is licensed by the US Food and Drug Administration for individuals 9 through 16 years of age with laboratory-confirmed previous dengue infection and who live in endemic areas [6]. Recently, Takeda Pharmaceuticals published the initial results of their tetravalent live-attenuated vaccine (TAK-003) in 20 071 participants through 1 year post vaccination [7]. The overall vaccine efficacy in the safety pop-ulation was 80.9% (95% confidence interval [CI], 75.2–85.3). However, wide ranging confidence intervals for dengue virus (DENV) serotypes 1 and 3 along with indeterminable efficacy for serotype 4 suggests tetravalent protection may not be in-duced by this vaccine.

All the current dengue vaccines in clinical trials are live-attenuated tetravalent vaccines requiring utilizing 1 or multiple doses (homologous prime-boost strategy) to achieve adequate tet-ravalent neutralizing antibody titers. A heterologous prime-boost strategy, which involves consecutive doses of different vaccine platforms, has the advantage of improving both humoral and cel-lular immunity to a specific antigen [8–10]. For dengue vaccines, a prime-boost immunization strategy has been proposed to induce

applyparastyle “fig//caption/p[1]” parastyle “FigCapt”

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

1708 • jid 2021:223 (15 May) • Lin et al

or boost a cell mediated immune response while avoiding the in-duction of antibodies that potentially mediate antibody-dependent enhancement [10]. This technique was employed in rhesus ma-caques by priming with a tetravalent purified inactivated dengue vaccine followed by boosting with a tetravalent live-attenuated dengue vaccine [11]. This strategy resulted in high titers of neu-tralizing antibody and protection upon challenge.

We describe the results of a phase 1 clinical trial testing het-erologous prime-boost vaccine administration strategies using different dosing schedules of tetravalent live and inactivated dengue vaccine candidates (ClinicalTrials.gov NCT02239614). The objective of the trial was to explore whether a heterologous prime-boost vaccination strategy could improve reactogenicity profiles, compress dosing schedules, and improve the breadth of immunologic responses when compared to historic ex-periments testing homologous prime-boost administration strategies.

METHODS

Study Objectives and Design

The primary study objectives were to evaluate the safety and immunogenicity of 4 different vaccine administration strategies from day 0 through 28 days after each dose. This study was a phase 1, randomized, open-label, study with 4 treatment groups consisting of 2 tetravalent dengue vaccine candidates adminis-tered using a heterologous prime-boost administration strategy and 2 dosing schedules (day 0 and day 28 vs day 0 and day 180). The study was conducted under an Investigational New Drug application filed with the US Food and Drug Administration. The study was conducted at the Walter Reed Army Institute of Research (WRAIR), Clinical Trials Center, Silver Spring, MD (ClinicalTrials.gov NCT02239614). The WRAIR Institutional Review Board and United States Army Medical Research and Materiel Command Office of Research Protections reviewed and approved the protocol. The investigators have adhered to the policies for protection of human subjects as prescribed in Army Regulation 70-25.

Vaccines

Two investigational products were studied, a tetravalent dengue virus (DENV-1–4) purified formalin-inactivated vaccine (TDENV-PIV) with alum adjuvant and a tetravalent dengue virus (DENV-1–4) live attenuated vaccine (TDENV-LAV) de-scribed previously [11–17]. TDENV-PIV consists of a tetrava-lent formulation of the following nonattenuated dengue virus strains: West Pac 74 (DENV-1), S16803 (DENV-2), CH53489 (DENV-3), and TVP360 (DENV-4), propagated in Vero cells, purified, inactivated with formalin, and formulated with 4 µg per DENV serotype per dose. TDENV-PIV was adjuvanted with alum (Alhydrogel 2%, 10.38  mg of Al3+/mL; Brenntag Biosector); after dilution, each 0.5-mL vaccine dose contained 500 μg of Al3+. The TDENV-LAV vaccine was made using the

following attenuated virus seeds: DENV-1 (45AZ5, PDK-27), DENV-2 (S16803, PDK-50), DENV-3 (CH53489, PDK-20), and DENV-4 (341750, PDK-6). Clinical vaccine lots were pre-pared at the WRAIR Pilot Bioproduction Facility, Silver Spring, MD. Each vaccine was delivered in a 0.5-mL dose volume using a needle and syringe; TDENV-PIV was administered intramus-cularly while TDENV-LAV was administered subcutaneously.

Study Volunteers and Enrollment

Healthy male and female adults between 18 and 49 years of age were recruited. Female participants had to be of nonchildbearing potential, abstinent, or had to use adequate contraceptive pre-cautions for 30 days prior to vaccination, had a negative preg-nancy test on the day of vaccination, and agreed to continue such precautions for 90 days after completion of the vaccina-tion series. Exclusion criteria were seropositivity for hepatitis B, hepatitis C, human immunodeficiency virus, or a history of chronic disease, chronic alcohol consumption and/or drug abuse, receipt of immunoglobulins and/or any blood products within 90  days preceding vaccination or planned administra-tion during the study period, as well as clinically significant laboratory abnormalities. Participants were not screened for fla-vivirus antibodies at enrollment but the according-to-protocol (ATP) analysis of immunogenicity included only participants who were seronegative for all 4 DENV serotypes at baseline (50% microneutralization assay end point [MN50] titer <1:10). Serostatus for DENV was determined after enrollment based on prevaccination blood samples.

A total of 80 volunteers were enrolled and randomized to 1 of 4 groups in a 1:1:1:1 distribution. Block randomization was used to allocate volunteers to each study group divided into 20 blocks of 4 by the order of study enrollment. Group 1 received a single dose of TDEN-LAV at day 0 and a boost of TDENV-PIV at day 28; group 2 received TDENV-PIV at day 0 and TDENV-LAV at day 28; group 3 received TENV-LAV at day 0 and TDENV-PIV at day 180; and group 4 received TDENV-PIV at day 0 and TDENV-LAV at day 180.

Safety Evaluation

Solicited injection site and general adverse events (AEs) were followed for 21 days following each dose using both a memory aid completed by the volunteer and clinical visits with an investigator. Unsolicited AEs were recorded for 28  days fol-lowing each dose and were coded with the use of the “Medical Dictionary for Regulatory Activities.” Serious adverse events (SAEs), potential immune-mediated diseases, and medically attended events were recorded throughout the entire study period. Each group was split into 2 subgroups (A or B) each corresponding to a blood collection schedule. Splitting the groups allowed greater breadth of sampling over time while reducing the overall blood donation burden for each indi-vidual volunteer.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

Dengue Prime-Boost Vaccines • jid 2021:223 (15 May) • 1709

Viremia

Assessment for viremia was conducted at WRAIR using quantitative reverse transcription polymerase chain reaction (RT-PCR) as previously described. Viremia after TDENV-LAV vaccination typically occurs 5–14 days post vaccination based on previous data [12, 13, 15, 16]. Viremia was not expected after TDENV-PIV vaccination. Viremia was measured at 5 time points per volunteer following TDENV-LAV.

Neutralizing Antibody Assay

DENV neutralizing antibodies were measured using a microneutralization assay to estimate MN50 as previously de-scribed [14–17]. The parental virus strains to be neutralized were DENV-1 WP74, DENV-2 S16803, DENV-3 CH53489, and DENV-4 341750. Seropositivity was defined as a titer ≥1:10.

Blood Sampling

Blood sampling is detailed in Supplementary Figure 1.

Immunogenicity Assessment

Cell-mediated immunity responses to vaccination were as-sessed by interferon-γ enzyme-linked immunospot (IFN-γ ELISPOT) assay prevaccination (study day 0)  and at 28  days and 180 days post dose 2 in all groups using methods as pre-viously described [18]. Briefly, cryopreserved peripheral blood mononuclear cells (PBMCs) were thawed and rested overnight before plating at 100 000–200 000 cells per well with 1 μg/mL/peptide of the applicable peptide pool or medium plus 0.5% DMSO (negative control). Peptide pools included overlapping peptides spanning the C/prM, E, NS1, NS3, and NS5 protein regions of all 4 DENV types, as previously described [18]. Data were normalized to spot-forming cells (SFC) per million PBMC for analysis and reporting purposes.

Statistical Analysis

The total vaccinated population includes all volunteers who received both vaccine doses. The ATP immunogenicity co-hort includes all volunteers who received both doses of study vaccine and were seronegative to all 4 DENV types at baseline. The percentages of volunteers reporting any AE within 28 days (0–27) following vaccination were computed by study group ac-cording to the type of AE, intensity, and relationship to vaccina-tion. The percentages of volunteers with measurable viremia by DENV type were tabulated after each dose and for each study group. Seropositivity rates for each DENV type and tetrava-lent antibody responses as determined by MN50 titer were cal-culated with 95% CI using Wald method for each vaccination group. Antibody titers were summarized as geometric mean titers (GMT), with the 95% CI, and reverse cumulative curves. A DENV-primed volunteer was defined as having a MN50 titer of ≥10 in the prevaccination (day 0) sample. Tetravalent sero-positivity rates were defined as the percentage of volunteers who were seropositive to all 4 DENV types. The GMT calculations

were performed by taking the antilog of the mean of the log 10 titer transformations. Antibody titers below the cutoff of the assay were given an arbitrary value of 5 (half the cutoff value) for the purpose of GMT calculation. For a given volunteer and a given immunogenicity measurement, missing or nonevaluable measurements were not replaced. Therefore, ATP analysis ex-cluded volunteers with missing or nonevaluable measurements.

RESULTS

Study Population

Eighty volunteers were enrolled in the study and divided among the 4 different groups as shown in Figure 1; 94% (75/80) of vo-lunteers received their second vaccination. Five volunteers were withdrawn prior to the second vaccination. The ATP cohort in-cluded 17 subjects in groups 1 and 2, 20 subjects in group 3, and 12 subjects in group 4. Two subjects were withdrawn from group 4 due to consistent failure to present for scheduled study visits. The mean age of volunteers enrolled in the study was 34.9 years (range 19 to 50). There were 28 women (35%) and 52 men (65%). Most volunteers were either Caucasian (40%) or African American (49%).

Safety and Reactogenicity

The frequencies of solicited local AEs and systemic AEs are pre-sented in Table 1 and Table 2. Injection site pain was generally higher following administration of PIV compared to LAV (0%–40% vs 0%–20%) across all groups. No grade 3 solicited local AEs occurred in any group with either vaccination. Overall, most solicited systemic AEs were mild or moderate. The most commonly reported systemic AEs were fatigue (15%–40% with LAV and 10%–20% with PIV) and headache (15%–40% with LAV and 5%–25% with PIV). Myalgias and arthralgias were also commonly reported with generally higher rates in groups 2 and 4 compared to groups 1 and 3 but not reaching statistical significance. No fevers were recorded following PIV adminis-tration, but 20% of volunteers in group 2 and 25% of volun-teers in group 4 reported fever following administration of LAV. No volunteers in groups 1 or 3 reported any fevers following LAV. A maculopapular rash was observed in 8 volunteers fol-lowing LAV vaccination. Three occurred following LAV prime (2 in group 1 and 1 in group 3) and 5 occurred following LAV boost (2 in group 2 and 3 in group 4). Several volunteers re-ported transient grade 3 solicited systemic AEs: abdominal pain and nausea/vomiting following PIV; headache, fever, nausea/vomiting, myalgia, arthralgia, and fatigue rarely reported fol-lowing LAV. There were no study-related SAEs. Related lab-oratory abnormalities occurred rarely and included grade 1 leukopenia in 1 volunteer in group 1, grade 2 neutropenia in 1 volunteer in group 3, grade 2 hyperbilirubinemia in 1 volun-teer in group 3, and grade 2 neutropenia and lymphopenia in 1 volunteer in group 4. All of these laboratory abnormalities were transient and resolved without sequelae.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

1710 • jid 2021:223 (15 May) • Lin et al

RNAemia

Levels of peripheral RNAemia detected per visit and per volun-teer by group are displayed in Supplementary Table 1. RNAemia was detected in all groups, with DENV-2 and DENV-4 the most frequently detected. Overall, DENV-2 was detected in 63% of positive samples, DENV-4 in 31% of samples, and DENV-1 in 6%. DENV-3 RNAemia was not detected in any sample at any of the tested time points. Detectable RNAemia occurred most fre-quently between days 7 and 14 post LAV vaccination, while days 10 and 11 were the 2 days with the highest detection rate. A sig-nificantly greater number of volunteers in group 4 had detect-able RNAemia compared to other groups (P = .023). However, all volunteers in group 4 had detectable RNAemia at only 1 time point while in group 1, 3 volunteers had detectable RNAemia at 2 time points. Group 2 similarly had 3 volunteers with detect-able RNAemia at 2 time points and group 3 had 1 volunteer with detectable RNAemia at 3 time points. An analysis of frequency of detectable RNAemia days by group using a Fisher exact test failed to demonstrate any significant difference (P = .547). Levels of RNAemia were higher for DENV-4 compared to DENV-2 across all groups (Supplementary Figure 2).

ImmunogenicityHumoral ImmunogenicitySeropositivity rates based on neutralizing antibody titers by group and measured at baseline and 28 and 180 days following booster vaccination are presented in Table 3. PIV priming was superior to LAV priming when measured by the percent of the cohort with tetravalent seroconversion following a complete vaccination series. PIV priming followed by LAV boost resulted in 100% tetravalent seroconversion at 28  days post vaccina-tion when LAV was administered either 28 or 180  days later. In contrast, only 35% of LAV recipients developed a tetrava-lent response when boosted with PIV at 28 days and 75% when boosted at 180 days. The distribution of neutralizing antibody responses across the 4 serotypes was also more balanced in the PIV primed groups versus the LAV primed groups. At 28 days following a LAV-PIV prime-boost regimen, DENV-1–4 sero-conversion rates were 71%, 82%, 41%, and 47% in the 0–28 day group and 90%, 90%, 85%, and 75% for the 0–180 day group. In contrast, the PIV-prime LAV-boost regimen resulted in se-roconversion rates of 100% for each serotype for both 0–28 and 0–180 dosing regimens. At 180 days following a LAV-PIV

Group 1 n = 20LAV/PIV 1 Month

Group 2 n = 20LAV/PIV 1 Month

Group 3 n = 20LAV/PIV 6 Month

Total Enrolled n = 80

20 receive 1st vaccination 20 receive 1st vaccination 20 receive 1st vaccination

1 withdrawal due to AE 1 withdrawn 0 withdrawals

19 receive 2nd vaccination 19 receive 2nd vaccination 20 receive 2nd vaccination

1 withdrawal 1 lost to follow up 0 withdrawals

18 completed study 18 completed study 20 completed study

1 dengue seropositive atbaseline

1 dengue seropositive atbaseline

0 dengue seropositive atbaseline

17 ATPimmunogenicity cohort

17 ATPimmunogenicity cohort

20 ATPimmunogenicity cohort

Group 4 n = 20PIV/LAV 6 Month

20 receive 1st vaccination

3 withdrawn (2 physiciandecision, 1 pregnancy)

17 receive 2nd vaccination

1 withdrawal and 1 lostto follow up

15 completed study

3 dengue seropositive atbaseline

12 ATPimmunogenicity cohort

Figure 1. Enrollment scheme: group 1, LAV day 0/PIV day 28; group 2, PIV day 0/LAV day 28; group 3, LAV day 0/PIV day 180; group 4, PIV day 0/LAV day 180. Abbreviations: ATP, according to protocol; LAV, live attenuated vaccine; PIV, purified inactivated vaccine.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

Dengue Prime-Boost Vaccines • jid 2021:223 (15 May) • 1711

prime-boost regimen, DENV-1–4 seroconversion rates were 47%, 77%, 41%, and 29% for the 0–28 day dosing and 60%, 80%, 45%, and 30% for the 0–180 day dosing. When the prime-boost regimen was PIV-LAV the seroconversion rates for DENV-1–4 were 94%, 94%, 88%, and 82% for the 0–28 dosing and 100% for all serotypes for the 0–180 regimen.

GMTs were also higher in PIV primed groups (Figure  2). When measured 28 days following the booster dose, LAV-PIV regimens had GMTs across the DENV-1–4 serotypes of 37, 280, 22, and 25 for the 0–28 dosing regimen and 107, 308, 89, and 48 for the 0–180 regimen. The PIV-LAV regimens had DENV-1–4 GMTs of 915, 2370, 772, and 1052 for the 0–28 dosing regimen and 3459, 4472, 4043, and 4141 for the 0–180 dosing regimen (Supplementary Table 2). These data indicated the PIV-prime LAV-boost regimen induced higher GMTs overall, and that this was more pronounced in the group that received a later boost (0–180) as compared to an earlier boost (0–28) as measured 180 days following the final vaccination.

Cell-Mediated ImmunogenicityResponse rates to vaccination as assessed using an IFN-γ ELISpot assay for each serotype are shown in Supplementary Table 3. Only DENV-2–derived peptides elicited response rates that were statistically greater than prevaccination for all groups at both 28  days and 180  days postvaccination. In all groups DENV-2–derived peptides were the most frequently recog-nized antigens at both time points post vaccination. There were 9 subjects (3 from group 1; 1 from group 2; 4 from group 3; and 1 from group 4) who demonstrated no ELISpot response post vaccination. However, there was no diminution of the response rates from days 28 to 180 post vaccination among responders. The valency of the response post vaccination (as determined by overlapping peptide pools representing the C/prM, E, NS1, NS3, and NS5 protein regions of each DENV type) was pre-dominantly multivalent with combined tri- and tetravalent

response rates ranging from 20% to 75% (Figure 3). There was no diminution of response valency from day 28 to day 180 post vaccination, with group 4 displaying the greatest breadth of response (75% tetravalent) at 180  days post vaccination. Response magnitudes for positive responder subjects are shown in Figure 4 and Supplementary Table 4. Responses were robust and frequently exceeded 1000 SFC/106 PBMC. For all peptide sets and all groups there was a significant increase in ELISpot response magnitude at both postvaccination time points com-pared to prevaccination.

DISCUSSION

A heterologous prime-boost vaccination strategy combining live and inactivated vaccine products has the potential to gen-erate durable humoral and cellular immune responses to each of the 4 dengue serotypes. Our results demonstrated that priming with TDEN-PIV and boosting with TDEN-LAV resulted in higher tetravalent seroconversion rates and higher neutralizing antibody titers than priming with TDEN-LAV and boosting with TDEN-PIV. Neutralizing antibody titers and seroconver-sion rates for the PIV followed by LAV prime-boost regimen exceeded those achieved following 2 doses of LAV or 2 doses of PIV with alum administration in previous studies [15, 19]. Two doses of PIV with ASO1E or ASO3B adjuvants significantly im-proved humoral responses compared to alum-adsorbed PIV; however, all 2-dose PIV regimens resulted in rapidly waning neutralizing antibody titers by 6 months post dose 2 [19].

In our study, PIV followed by LAV at 1  month generated significantly higher titers compared to the best performing PIV adjuvanted 2-dose homologous prime-boost vaccina-tion. Spacing the LAV booster dose to 6 months following PIV priming further increased the humoral response in magnitude. Future trials will address whether a schedule in between 1 and 6 months will induce a comparable antibody profile while min-imizing the time between doses. Additionally, human challenge

Table 1. Incidence of Solicited Local Symptoms During the 21 Days Following Each Vaccination

Symptom Vaccine Grade

Group 1 (n = 20) Group 2 (n = 20) Group 3 (n = 20) Group 4 (n = 20)

n (%) 95% CI n (%) 95% CI n (%) 95% CI n (%) 95% CI

Pain LAV Any 1 (5.0) .1–24.9 2 (10.0) 1.2–31.7 4 (20.0) 5.7–43.7 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

PIV Any 3 (15.0) 3.2–37.9 5 (25.0) 8.7–49.1 7 (35.0) 15.4–59.2 8 (40.0) 19.1–63.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Redness LAV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8 1 (5.0) .1–24.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

PIV Any 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Swelling LAV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8 1 (5.0) .1–24.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

PIV Any 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Abbreviations: CI, confidence interval; LAV, live attenuated vaccine; PIV, purified inactivated vaccine.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

1712 • jid 2021:223 (15 May) • Lin et al

studies are planned to address the potential for protective efficacy.

IFN-γ–mediated T-cell responses induced by the 4 different vaccination regimens tested in this study were variable overall, but comparable among the 4 groups. Each group contained some very high responders (>1000 SFC/106 PBMC) for all sero-types and some nonresponders (<38 SFC/106 PBMC) against any serotype. DENV-2 was the most frequently recognized se-rotype and induced the highest magnitude IFN-γ response in all groups at both time points. This observation is consistent with the RNAemia data and the notion that T-cell immunity is

driven by actively replicating vaccine virus. Importantly, T-cell responses did not wane significantly in frequency or magnitude up to 180  days post vaccination. Of the 4 groups, group 4 in particular induced T-cell responses that best met 3 important qualities of a DENV vaccine: balanced, durable, and tetravalent.

Overall, the heterologous prime-boost strategy as utilized in this study resulted in an acceptable safety profile. In general, it was noted that there were higher rates of multiple solicited sys-temic AEs in the PIV primed groups along with a notably higher rate of grade 3 solicited systemic AEs in group 4.  It was also noted that unprimed LAV recipients had no recorded fevers, in

Table 2. Incidence of Solicited Systemic Adverse Events During the 21 Days Following Each Vaccination

Symptom Vaccine Grade

Group 1 (n = 20) Group 2 (n = 20) Group 3 (n = 20) Group 4 (n = 20)

n (%) 95% CI n (%) 95% CI n (%) 95% CI n (%) 95% CI

Abdominal Pain LAV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 2 (10.0) 1.2–31.7 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

PIV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Arthralgia LAV Any 2 (10.0) 1.2–31.7 5 (25.0) 8.7–49.1 2 (10.0) 1.2–31.7 4 (20.0) 5.7–43.7

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9

PIV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 2 (10.0) 1.2–31.7 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Diarrhea LAV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 1 (5.0) .1–24.9 1 (5.0) .1–24.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

PIV Any 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Fatigue LAV Any 3 (15.0) 3.2–37.9 8 (40.0) 19.1–63.9 5 (25.0) 8.7–49.1 5 (25.0) 8.7–49.1

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 2 (10.0) 1.2–31.7

PIV Any 2 (10.0) 1.2–31.7 2 (10.0) 1.2–31.7 3 (15.0) 3.2–37.9 4 (20.0) 5.7–43.7

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Fever LAV Any 0 (0.0) .0–16.8 5 (25.0) 8.7–49.1 0 (0.0) .0–16.8 5 (25.0) 8.7–49.1

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9

PIV Any 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Headache LAV Any 3 (15.0) 3.2–37.9 8 (40.0) 19.1–63.9 8 (40.0) 19.1–63.9 6 (30.0) 11.9–54.3

Grade 3 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8 2 (10.0) 1.2–31.7

PIV Any 1 (5.0) .1–24.9 3 (15.0) 3.2–37.9 5 (25.0) 8.7–49.1 3 (15.0) 3.2–37.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Myalgia LAV Any 1 (5.0) .1–24.9 5 (25.0) 8.7–49.1 2 (10.0) 1.2–31.7 6 (30.0) 11.9–54.3

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9

PIV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 4 (20.0) 5.7–43.7 3 (15.0) 3.2–37.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8 0 (0.0) .0–16.8

Nausea LAV Any 1 (5.0) .1–24.9 1 (5.0) .1–24.9 3 (15.0) 3.2–37.9 1 (5.0) .1–24.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8

PIV Any 0 (0.0) .0–16.8 1 (5.0) .1–24.9 1 (5.0) .1–24.9 1 (5.0) .1–24.9

Grade 3 0 (0.0) .0–16.8 1 (5.0) .1–24.9 1 (5.0) .1–24.9 0 (0.0) .0–16.8

Vomiting LAV Any 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9 1 (5.0) .1–24.9

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8

PIV Any 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8

Grade 3 0 (0.0) .0–16.8 0 (0.0) .0–16.8 1 (5.0) .1–24.9 0 (0.0) .0–16.8

Overall LAV Any 5 (25.0) 8.7–49.1 10 (50.0) 27.2–72.8 8 (40.0) 19.1–63.9 8 (40.0) 19.1–63.9

Grade 3 0 (0.0) .0–16.8 1 (5.0) .1–24.9 1 (5.0) .1–24.9 4 (20.0) 5.7–43.7

PIV Any 2 (10.0) 1.2–31.7 4 (20.0) 5.7–43.7 5 (25.0) 8.7–49.1 8 (40.0) 19.1–63.9

Grade 3 0 (0.0) .0–16.8 1 (5.0) .1–24.9 1 (5.0) .1–24.9 0 (0.0) .0–16.8

Abbreviations: CI, confidence interval; LAV, live attenuated vaccine; PIV, purified inactivated vaccine.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

Dengue Prime-Boost Vaccines • jid 2021:223 (15 May) • 1713

Table 3. Seropositivity Rates by Group at Baseline and 28 and 180 Days Following Booster Vaccination

Serotype/Time point

Group 1 (n = 17) Group 2 (n = 17) Group 3 (n = 20) Group 4 (n = 12)

n (%) 95% CI n (%) 95% CI n (%) 95% CI n (%) 95% CI

DENV-1

Baseline 0 (0.0) .0–19.5 0 (0.0) .0–19.5 0 (0.0) .0–16.8 0 (0.0) .0–26.5

28 days 12 (70.6) 44.0–89.7 17 (100.0) 80.5–1.0 18 (90.0) 68.3–98.8 12 (100.0) 73.5–1.0

180 days 8 (47.1) 23.0–72.2 16 (94.1) 71.3–99.9 12 (60.0) 36.1–80.9 12 (100.0) 73.5–1.0

DENV-2

Baseline 0 (0.0) .0–19.5 0 (0.0) .0–19.5 0 (0.0) .0–16.8 0 (0.0) .0–26.5

28 days 14 (82.4) 56.6–96.2 17 (100.0) 80.5–1.0 18 (90.0) 68.3–98.8 12 (100.0) 73.5–1.0

180 days 13 (76.5) 50.1–93.2 16 (94.1) 71.3–99.9 16 (80.0) 56.3–94.3 12 (100.0) 73.5–1.0

DENV-3

Baseline 0 (0.0) .0–19.5 0 (0.0) .0–19.5 0 (0.0) .0–16.8 0 (0.0) .0–26.5

28 days 7 (41.2) 18.4–67.1 17 (100.0) 80.5–1.0 17 (85.0) 62.1–96.8 12 (100.0) 73.5–1.0

180 days 7 (41.2) 18.4–67.1 15 (88.2) 63.6–98.5 9 (45.0) 23.1–68.5 12 (100.0) 73.5–1.0

DENV-4

Baseline 0 (0.0) .0–19.5 0 (0.0) .0–19.5 0 (0.0) .0–16.8 0 (0.0) .0–26.5

28 days 8 (47.1) 23.0–72.2 17 (100.0) 80.5–1.0 15 (75.0) 50.9–91.3 12 (100.0) 73.5–1.0

180 days 5 (29.4) 10.3–56.0 14 (82.4) 56.6–96.2 6 (30.0) 11.9–54.3 12 (100.0) 73.5–1.0

Tetravalent seropositivity

Baseline 0 (0.0) .0–19.5 0 (0.0) .0–19.5 0 (0.0) .0–16.8 0 (0.0) .0–26.5

28 days 6 (35.3) 14.2–61.7 17 (100.0) 80.5–1.0 15 (75.0) 50.9–91.3 12 (100.0) 73.5–1.0

180 days 5 (29.4) 10.3–56.0 12 (70.6) 44.0–89.7 5 (25.0) 8.7–49.1 12 (100.0) 73.5–1.0

Abbreviations: CI, confidence interval; DENV, dengue virus.

100 0001 month

DENV-1

6 months

10 000

1000

MN

50 ti

ter

100

10

1G1 G2 G3 G4 G1 G2 G3 G4

100 0001 month

DENV-2

6 months

10 000

1000

MN

50 ti

ter

100

10

1G1 G2 G3 G4 G1 G2 G3 G4

100 0001 month

DENV-3

6 months

10 000

1000

MN

50 ti

ter

100

10

1G1 G2 G3 G4 G1 G2 G3 G4

100 0001 month

DENV-4

6 months

10 000

1000

MN

50 ti

ter

100

10

1G1 G2 G3 G4 G1 G2 G3 G4

Figure 2. Neutralizing antibody titers by group, serotype, and measurement at 28 and 180 days following booster vaccination. Abbreviations: DENV, dengue virus; G1, group 1; G2, group 2; G3, group 3; G4, group 4; MN50, 50% microneutralization end point.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

1714 • jid 2021:223 (15 May) • Lin et al

100 000

1000

SFC

/106 P

BM

C

100

1 2 3 4 1 2 3 4 1 2 3 4

Day 180Post-vacc

Day 28Post-vacc

Post-vacc

100 000

1000

SFC

/106 P

BM

C

100

1 2 3 4 1 2 3 4 1 2 3 4

Day 180Post-vacc

Day 28Post-vacc

Post-vacc

100 000

Group 3 Group 4

Group 1A B

C D

Group 2

1000

SFC

/106 P

BM

C

100

1 2 3 4 1 2 3 4 1 2 3 4

Day 180Post-vacc

Day 28Post-vacc

Post-vacc

100 000

1000

SFC

/106 P

BM

C

100

1 2 3 4 1 2 3 4 1 2 3 4

Day 180Post-vacc

Day 28Post-vacc

Post-vacc

Figure 4. Total ELISpot response frequencies. Each serotype (1 through 4) is designated for each time point tested. Total frequencies were calculated by summing the SFC/106 PBMC value for each of the 5 peptide pools matching a given serotype (C/preM, E, NS1, NS3, and NS5). Each dot represents the response frequency of a single sub-ject to a given serotype. Abbreviations: ELISpot, enzyme-linked immunospot; PBMC, peripheral blood mononuclear cells; Postvacc, postvaccination; Prevacc, prevaccination; SFC, spot-forming cells.

Prevacc

Group 1LAV/PIV(28)

Group 2PIV/LAV(28)

Group 3LAV/PIV(180)

Group 4PIV/LAV(180)

Negative

Monovalent

Bivalent

Trivalent

Tetravalent

Day 28post

dose 2

Day 180Post-

Dose 2

Figure 3. ELISpot response valency. Pie chart segments represent the fraction of subjects responding to at least 1 peptide pool from 0, 1, 2, 3, or 4 DENV serotypes. Abbreviations: ELISpot, enzyme-linked immunospot; DENV, dengue virus; LAV, live attenuated vaccine; PIV, purified inactivated vaccine; Prevacc, prevaccination.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

Dengue Prime-Boost Vaccines • jid 2021:223 (15 May) • 1715

contrast to PIV-primed LAV recipients. This observation stands in contrast to previous studies with this same TDEN-LAV for-mulation, wherein recorded fever rates of 9.1%–18.2% were observed [15]. Overall, given the small numbers available from this phase 1 study, confidence intervals between the observed rates of these AEs overlap significantly suggesting that further study is warranted to confirm whether or not reactogenicity is, in fact, increased by PIV priming or that this differential rate was observed by chance alone.

The mechanisms underlying the improvement in antibody titers along with the trend towards greater reactogenicity ob-served in the PIV/LAV groups are not yet fully characterized by this study. The antibody titers generated by 1 dose of PIV alone were overall very low or undetectable and so instead of neu-tralizing vaccine strains from the LAV, it is possible that these low levels of antibody actually enhanced vaccine strain replica-tion through an antibody-dependent enhancement mechanism. Further studies to evaluate the immune mechanisms driving the differential responses observed in this study are warranted and currently ongoing.

This study had a number of limitations. The open-label de-sign may have impacted the unbiased assessment of AEs. The vaccines have distinct appearances and are administered dif-ferently (intramuscular vs subcutaneous), making a double-blinded study design difficult. There was also an unexpectedly high number of subject withdrawals, particularly in group 4, which were unrelated to study events. Lastly, sampling of RNAemia at a limited number of time points on an alternating day schedule may have resulted in missed detection for some subjects with viremia on 1 day only.

In summary, this first-in-human dengue heterologous prime-boost trial had an acceptable safety profile. This trial established the superior humoral immunogenicity of priming with an in-activated vaccine candidate followed by an attenuated live virus vaccine. This trial supports continued testing of a heterologous prime-boost strategy against dengue.

Supplementary Data

Supplementary materials are available at The Journal of Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or com-ments should be addressed to the corresponding author.

Notes

Acknowledgments. We acknowledge the staff of the Walter Reed Army Institute of Research (WRAIR) Clinical Trial Center, the WRAIR Quality Management Unit, and spec-imen processing teams, along with all of the study volunteers who made this study possible. The study was sponsored by the Surgeon General, Department of the Army. The US Army Medical Materiel Development Activity, as the Office of the

Surgeon General’s sponsor representative, monitored and re-ported on volunteer safety.

Disclaimer. Material has been reviewed by the WRAIR. There is no objection to its presentation and/or publication. The opin-ions or assertions contained herein are the private views of the authors, and are not to be construed as official, or as reflecting true views of the Department of the Army or the Department of Defense. All authors have reviewed the manuscript and vouch for its accuracy and completeness.

Financial support. This work was supported by the Military Infectious Diseases Research Program.

Potential conflicts of interest. S. J. T. has been compensated as an advisory board member for Sanofi-Pasteur, Takeda, Merck, PrimeVax, Chugai, Tremau, and Janssen; and is a patent holder on combination flavivirus vaccine strategy patent number US10086061B2. K. H. E.  is a patent holder on the tetravalent live attenuated dengue vaccine and tetravalent purified inacti-vated vaccine products utilized in this study. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

1. Bhatt S, Gething PW, Brady OJ, et al The global distribution and burden of dengue. Nature 2013; 496:504–7.

2. Prompetchara  E, Ketloy  C, Thomas  SJ, Ruxrungtham  K. Dengue vaccine: global development update [published on-line ahead of print 13 January  2019]. Asian Pac J Allergy Immunol doi: 10.12932/AP-100518-0309.

3. Deng  SQ, Yang  X, Wei  Y, Chen  JT, Wang  XJ, Peng  HJ. A review on dengue vaccine development. Vaccines (Basel) 2020; 8:63.

4. Torresi  J, Ebert  G, Pellegrini  M. Vaccines licensed and in clinical trials for the prevention of dengue. Hum Vaccin Immunother 2017; 13:1059–72.

5. Thomas SJ. The necessity and quandaries of dengue vaccine development. J Infect Dis 2011; 203:299–303.

6. US Food and Drug Administration. Dengvaxia. https://www.fda.gov/vaccines-blood-biologics/dengvaxia. Accessed 2 June 2020.

7. Biswal  S, Reynales  H, Saez-Llorens  X, et  al; TIDES Study Group. Efficacy of a tetravalent dengue vaccine in healthy children and adolescents. N Engl J Med 2019; 381:2009–19.

8. Kardani K, Bolhassani A, Shahbazi S. Prime-boost vaccine strategy against viral infections: mechanisms and benefits. Vaccine 2016; 34:413–23.

9. Lu  S. Heterologous prime-boost vaccination. Curr Opin Immunol 2009; 21:346–51.

10. Valdés  I, Lazo L, Hermida L, Guillén G, Gil L. Can com-plementary prime-boost immunization strategies be an

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022

1716 • jid 2021:223 (15 May) • Lin et al

alternative and promising vaccine approach against dengue virus? Front Immunol 2019; 10:1956.

11. Simmons  M, Burgess  T, Lynch  J, Putnak  R. Protection against dengue virus by non-replicating and live attenu-ated vaccines used together in a prime boost vaccination strategy. Virology 2010; 396:280–8.

12. Watanaveeradej  V, Simasathien  S, Mammen  MP, et  al Long-term safety and immunogenicity of a tetravalent live-attenuated dengue vaccine and evaluation of a booster dose administered to healthy Thai children. Am J Trop Med Hyg 2016; 94:1348–58.

13. Watanaveeradej V, Gibbons RV, Simasathien S, et al Safety and immunogenicity of a rederived, live-attenuated dengue virus vaccine in healthy adults living in Thailand: a ran-domized trial. Am J Trop Med Hyg 2014; 91:119–28.

14. Martinez LJ, Lin L, Blaylock JM, et al Safety and immunogenicity of a dengue virus serotype-1 purified-inactivated vaccine: results of a phase 1 clinical trial. Am J Trop Med Hyg 2015; 93:454–60.

15. Thomas SJ, Eckels KH, Carletti I, et al A phase II, random-ized, safety and immunogenicity study of a re-derived,

live-attenuated dengue virus vaccine in healthy adults. Am J Trop Med Hyg 2013; 88:73–88.

16. Bauer  K, Esquilin  IO, Cornier  AS, et  al A phase II, ran-domized, safety and immunogenicity trial of a re-derived, live-attenuated dengue virus vaccine in healthy children and adults living in Puerto Rico. Am J Trop Med Hyg 2015; 93:441–53.

17. Fernandez  S, Thomas  SJ, De  La  Barrera  R, et  al An adjuvanted, tetravalent dengue virus purified inactivated vaccine candidate induces long-lasting and protective an-tibody responses against dengue challenge in Rhesus ma-caques. Am J Trop Med Hyg 2015; 92:698–708.

18. Waickman AT, Friberg H, Gargulak M, et al Assessing the diversity and stability of cellular immunity generated in re-sponse to the candidate live-attenuated dengue virus vac-cine TAK-003. Front Immunol 2019; 10:1778.

19. Schmidt AC, Lin L, Martinez LJ, et al Phase 1 randomized study of a tetravalent dengue purified inactivated vaccine in healthy adults in the United States. Am J Trop Med Hyg 2017; 96:1325–37.

Dow

nloaded from https://academ

ic.oup.com/jid/article/223/10/1707/5910633 by guest on 27 M

ay 2022