DENGUE40506 2023 Article 263
DENGUE40506 2023 Article 263
DENGUE40506 2023 Article 263
DOI 10.1007/s40506-023-00263-w
Keywords Dengue fever · Vascular leakage · Therapeutics · Vaccines · Metabolic disorders · Mast cells
Opinion statement
Dengue viruses (DENV) continue to circulate worldwide, resulting in a significant burden
on human health. There are four antigenically distinct serotypes of DENV, an infection
of which could result in a potentially life-threatening disease. Current treatment options
are limited and rely on supportive care. Although one dengue vaccine is approved for
dengue-immune individuals and has modest efficacy, there is still a need for therapeutics
and vaccines that can reduce dengue morbidities and lower the infection burden. There
have been recent advances in the development of promising drugs for the treatment of
dengue. These include direct antivirals that can reduce virus replication as well as host-
targeted drugs for reducing inflammation and/or vascular pathologies. There are also new
vaccine candidates that are being evaluated for their safety and efficacy in preventing
dengue disease. This review highlights nuances in the current standard-of-care treatment
of dengue. We also discuss emerging treatment options, therapeutic drugs, and vaccines
that are currently being pursued at various stages of preclinical and clinical development.
Vol.:(0123456789)
28
Introduction
Dengue fever (DF), caused by the dengue virus in the forest [4, 5] while in the urban cycle of transmis-
(DENV), is the world’s most prevalent and impor- sion, the virus is maintained within human popula-
tant arboviral infection. It is estimated that nearly tion aided by urban dwelling mosquitos such as Aedes
390 million infections occur annually, of which 96 aegypti [6]. As the geographical distributions of these
million manifest clinically [1]. DENV belongs to the vectors are expanding it is likely that DENV will spread
genus Flavivirus, which also comprises several other further [7]. There are 4 serotypes of DENV (DENV1-
clinically important human pathogens, such as Zika, 4), which makes it likely that an individual will be
Japanese encephalitis, West Nile, and Yellow Fever exposed to the virus multiple times in their lifetime
viruses, among others. The DENV genome consists [8]. Latest models of dengue transmission estimate
of a single-stranded positive-sense RNA that encodes that 4 million cases require hospitalization each year
for three structural (capsid, prM/M, and E) and seven [9] and account for an annual estimated cost of US
non-structural proteins that are translated during the $8.9 billion globally [10]. With an increase in urbani-
virus replication cycle [2]. The virus is spread by an zation and climate change supporting the spread of
infected mosquito bite during a blood meal and exists the mosquito vector, some models predict an increas-
in both sylvatic and urban ecosystems [3]. The sylvatic ing risk of DENV transmission, potentially impacting
cycle of DENV involves virus transmission between 6.1 billion people by 2080 [11].
non-human primates (NHP) and mosquitos prevalent
Fig. 1 The clinical management and possible treatment options for dengue disease. Diagram was made using biorender.com
JNJ-64281802 NS4B inhibitor that inhibits viral replication Antiviral activity in vitro was shown for its Clinical trials for dengue prophylaxis in
analog, JNJ-A07. Decrease in viremia, viral healthy individuals (NCT05201794) as well
burden, and inflammatory cytokines, and as for dengue therapy in patients with
improved survival in immunocompromised confirmed dengue fever (NCT04906980)
mouse model of DENV infection [53•] are in progress
Ivermectin Antiviral agent that inhibits host nuclear Reduced viral replication in vitro [56, 57] Randomized double-blind placebo-con-
import receptors trolled trial (n = 203) showed faster NS1
antigenemia clearance but no improve-
ment in virological or clinical efficacy
(NCT02045069) [58•]
AT-752 Guanosine nucleotide analog inhibiting NS5 Reduced viremia on certain days and Phase 1 (NCT05366439) and phase 2
RdRp function, inhibits viral replication improved survival in immunocompromised (NCT05466240) clinical trials are in
mice models infected with DENV [63] progress
Doxycycline Tetracycline class antibiotic, inhibits viral Antiviral activity against all four DENV One randomized clinical trial (n = 231)
entry and replication by inhibiting NS2B- serotypes in vitro [64] showed reduction in inflammatory
NS3 cytokines and another case control study
(n = 120) showed faster platelet recovery,
leukocyte count, and reduced length of
hospitalization. Clinical trials to test effi-
cacy of doxycycline as a dengue therapeu-
tic in pediatric (CTRI/2018/01/011548)
and adult (CTRI/2021/09/036661) popu-
lations are on-going
Dengue: Update on Clinically Relevant Therapeutic Strategies and Vaccines
Zanamivir Neuraminidase inhibitor to block desialyla- Reduction in DENV2 NS1-induced endothelial Clinical trial to test efficacy against vascular
tion on platelet membrane hyperpermeability and vascular leakage leakage (NCT04597437) is currently on-going
in vitro [73]
33
Table 1. (continued)
34
analog and is incorporated into RNA by RdRp, therein inhibiting viral replica-
tion. Correspondingly, AT-281, the prodrug of AT-752 inhibited DENV2 and
DENV3 in vitro and AT-752 showed reduced viremia (albeit only at day 6 and
8 post infection) and improved survival in DENV2-infected immunocompro-
mised mice. Currently, the drugs are in phase 1 (NCT05366439) and phase
2 (NCT05466240) double-blind, randomized, placebo-controlled studies to
assess the safety and antiviral activity in the dengue human challenge model
and in dengue patients, respectively.
Doxycycline, a broad-spectrum tetracycline-class antibiotic and antima-
larial, has shown some efficacy as an antiviral against DENV1-4 in vitro by
inhibiting NS2B-NS3 protease activity, resulting in reduced viral entry and
replication [64]. A randomized clinical trial (with no placebo control arm)
in Brazil, testing doxycycline for its efficacy as an anti-inflammatory drug
in dengue infection showed reduction in pro-inflammatory cytokines IL-6
and TNF in the treatment group compared to patients receiving standard
symptomatic and supportive care [65]. A case–control study in India showed
that doxycycline-treated dengue patients showed faster recovery of platelet
and leukocyte counts and reduced hospital stay [66]. However, these studies
so far have not been robust double-blind randomized placebo-controlled
clinical trials with defined end point measurements. One such clinical trial
(CTRI/2018/01/011548) is currently registered in India to study the efficacy of
doxycycline as a dengue antiviral in a pediatric population and could clarify
whether doxycycline has use as dengue therapeutic.
DENV infection and replication also require host factors that could be
potential drug targets. N-linked glycosylation of viral proteins (pre-membrane
(prM) and envelope (E) proteins) mediated by host α- glucosidase in the
endoplasmic reticulum is important for viral assembly and subsequent release
of mature, infectious DENV particles [67]. Celgosivir, an alpha-glucosidase
inhibitor, had impaired folding and trapping of NS1 and significant antivi-
ral activity when tested in vivo in mouse models of dengue infection [68].
Although the previous trial did not meet its primary outcome of reducing
viremia or fever, subsequent evaluation of the pharmacokinetic endpoints
of the trial inferred that a revised dosing regimen that would increase the
steady-state trough concentrations of the drug in the patient serum might be
more efficacious [52, 69]. UV-4B (N-(9′-methoxynonyl)-1-deoxynojirimycin),
another alpha-glucosidase inhibitor, also showed antiviral activity against
all four serotypes of dengue in vitro [70]. In vivo studies using immunosup-
pressed mice showed decreased viremia and viral burden and improved sur-
vival in a lethal ADE model of dengue disease [71]. A Phase 1a clinical trial
(NCT02061358) with healthy subjects, evaluating the safety, tolerability, and
pharmacokinetics of the drug, indicated that a single dose of up to 1000 mg
UV-4B was safe and well tolerated [72]. The efficacy of this drug in dengue
patients has yet to be determined.
Similar to oseltamivir mentioned earlier, Zanamivir, another neurami-
nidase inhibitor, has shown inhibition of DENV2 NS1-induced endothelial
hyperpermeability in vitro by inhibiting endogenous sialidase [73]. Consider-
ing that the TOTO trial failed to show any clinical benefit, the efficacy of zan-
amivir in vivo remains speculative. A phase 1 pilot randomized, double-blind,
placebo-controlled clinical trial for the evaluation of the safety and efficacy
38
Monoclonal therapeutics
An obstacle for the development of monoclonal antibody (mAb) thera-
peutics against DENV has been the identification of antibodies capable
of cross-neutralizing DENV1-4, which make up a minority of antibod-
ies induced by natural infection [78]. Many of the dominant antibod-
ies following natural infection target the immunodominant epitopes of
the E protein fusion loop and the prM proteins, which can be involved
in ADE [79]. Most mAb development pipelines have focused on the
goals of either developing broadly cross-neutralizing antibodies or pro-
ducing antibody cocktails [80]. Cross-reactive neutralizing antibodies
can be formed against domains I, II, and III of the E protein, yet those
against the quaternary structure of E protein dimers are more likely to be
potently cross-neutralizing [78, 79]. However, even highly potent neu-
tralizing antibodies can induce ADE at intermediate concentrations [8,
81], emphasizing the importance of safety testing of mAbs to identify
the optimal therapeutic window and concentration. The risk of ADE can
also be mitigated by engineering LALA substitutions in the heavy chain
[82, 83]. Monovalent mAb therapeutics also have the risk of becoming
obsolete if viral immune evasion results in loss of neutralizing epitopes,
as has been shown in the real world during the SARS-CoV-2 pandemic
[84], and in animal models of DENV mAb testing [82]. Development of
mAb as therapeutics or prophylactics for dengue has also been compre-
hensively reviewed elsewhere [85, 86] and three candidates for human
use are currently in clinical trials.
VIS513, a humanized pan-serotype anti-DENV mAb developed by Vis-
terra (Cambridge, Massachusetts) has been engineered to bind domain III
of the E protein of all 4 DENV serotypes [87]. Administration of VIS513
either 24 h or 5 days post dengue infection in non-human primates was
found to diminish infectious DENV in circulation without altering the
endogenous antibody response [88]. In the murine model of antibody-
enhanced DENV infection, VIS513 had increased protection of immu-
nocompromised mice from lethal primary and secondary infection [87].
VIS513 is currently in phase 2, a single-blind, randomized, parallel-group,
dose-ranging, single-dose study in India to study its safety and efficacy
in adults with dengue fever (CTRI/2021/07/035290). AV-1, developed
by AbViro LLC (NCT04273217), and Dengushield, developed by Serum
Institute of India Pvt. Ltd. (NCT03883620) are other human monoclonal
antibodies currently in phase 1, placebo-controlled, clinical trials with
Dengue: Update on Clinically Relevant Therapeutic Strategies and Vaccines Palanichamy Kala et al. 39
Vaccine development
Given the lack of specific dengue therapeutics and significant economic
and public health burden, WHO considers dengue vaccine development
an urgent priority [121]. Two main challenges prevail in dengue vaccine
development. Firstly, the vaccine should confer effective and balanced pro-
tection against all four dengue serotypes. Tetravalent vaccine formulations
developed with this aim are hindered by antigenic competition, wherein
the immune response is skewed towards more immunodominant serotype-
specific dengue antigens. Secondly, ADE poses a significant challenge in
dengue vaccine development since vaccine-mediated sub-optimal protec-
tion to any of the dengue serotypes can confer a risk of severe dengue in
a subsequent infection. Currently, there are at least seven dengue vaccines
42
the recent seroprevalence rate in the area has been documented to be at least
80% by 9 years of age [135].
QDENGA® or TAK-003 by Takeda Pharmaceuticals is another live-attenuated
tetravalent dengue vaccine that has been recently licensed for use in Indonesia
for individuals aged 6 to 45 [136], and has received marketing authorization
in the European Union for individuals aged 4 and above [137], regardless of
their prior dengue exposure status. It is currently undergoing regulatory review
in other dengue-endemic countries in Asia and Latin America [138, 139]. Using
attenuated DENV2 PDK-53 virus as backbone, the other chimeric dengue virus
were engineered by substituting the prM and E genes of DENV2 for that of
wild-type DENV1 16007, DENV3 16562, or DENV4 1036 virus [140]. It is
administered subcutaneously as a 0.5 mL dose in a two dose regimen scheduled
3 months apart [136, 137]. The approval is based on the Tetravalent Immuni-
zation against Dengue Efficacy Study (TIDES) trial which included more than
28,000 participants. The primary endpoint of the study was the overall vac-
cine efficacy against VCD in the first 11 months and was found to be 80·2%
(95% CI: 73·3–85·3) [141]. However, subsequent follow-up studies at 18-,
24- and 36-months vaccination showed waning of vaccine efficacy over time
with cumulative vaccine efficacy against VCD at 62% (95% CI: 56.6–66.7)
[123•, 124–143]. Nonetheless, vaccine efficacy against hospitalized VCD was
more robust and maintained from 90.4% (95% CI: 82.6–94.7) at 18 months
to 83.6% (95% CI: 76.8–88.4) at 3 years after the second dose. Importantly, the
year 3 cumulative data indicated no age effect and showed comparable overall
vaccine efficacy against VCD (65% vs 54.3%) and hospitalized VCD (86% vs
77.1%) for seropositive and seronegative individuals. When cumulative vaccine
efficacy was analyzed for specific serotypes, similar vaccine efficacy regardless
of baseline serostatus was observed for DENV-1 (seropositive: 56.2% (95% CI:
43.7–66.0) vs seronegative: 43.5% (95% CI: 21.5–59.3)) and DENV2 (seroposi-
tive: 83.4% (95% CI: 76.4–88.3) vs seronegative: 91.9% (95% CI: 83.6–96.0)).
However, while vaccine efficacy was 52.3% (95% CI: 36.6–64.2) for DENV-3
and 60.7% (95% CI: 16.0–81.6) for DENV-4 in seropositive individuals, no
efficacy was observed for DENV-3 (− 23.4% (95% CI: − 125.3–32.4)) and effi-
cacy against DENV-4 was inconclusive in seronegative patients [123•]. Notably
seronegative patients showed higher hospitalization rate (0.2%) compared to
placebo (< 0.1%) for DENV-3 infection [123•], raising concerns that serostatus
might also affect safety outcomes with this vaccine, similarly to Dengvaxia [144].
While this relative risk was inconclusive and was attributed by the authors to the
small number of DENV-3 cases and differences in clinical practice geographi-
cally, further analyses are warranted to ensure no increased hazard risk due to
ADE in seronegative patients. Moreover, the gradual waning efficacy needs to be
addressed. A new clinical trial (NCT03999996) to assess the impact of a booster
dose 15- and 24-months post 1 st dose of vaccine is currently on-going. In addi-
tion, the presence of non-structural (NS) proteins from the DENV-2 backbone
in Qdenga was shown to elicit NS-proteins specific T-cell mediated immunity
in samples from phase 2 trial [145], however, it is notable that the protection
offered by this vaccine is highest against DENV2, the strain from which the virus
backbone originates. This might point to the importance of ensuring that future
DENV vaccine development prioritizes generating tetravalent immunity with
respect to both antibodies and T cell epitopes.
44
Conclusion
The current standard of care for dengue remains supportive care. Timely fluid
management has shown to be effective in lowering deaths due to severe dengue.
However, due to the late onset of warning signs, it can be difficult to manage
severe dengue solely guided by symptomatic interventions and is often too late
too little for the interventions to be effective. Therefore, there is a dire need for
effective drugs that can reduce dengue viral infection and reduce the likelihood
of developing severe disease. Moreover, an integrated approach utilizing both
efficacious vaccines and drugs will be required to combat dengue globally. As
Dengue: Update on Clinically Relevant Therapeutic Strategies and Vaccines Palanichamy Kala et al. 45
highlighted in this review, there are new and promising drugs and vaccines for
the control of dengue at various stages of clinical development.
Funding
The authors acknowledge funding from the National Medical Research Council of Singapore (OFIR-
G22jul-0018 ) and National Research Foundation of Singapore (NRF-CRP27-2021-0003).
Declarations
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
Conflict of Interest
Dr. Rathore has patents related to dengue therapeutics, PCT/US11426408 and PCT/US11219616.
Dr. St. John has patents related to dengue therapeutics, PCT/US11426408 and PCT/US9730921B2.
Ms. Monica Palanichamy Kala declares no competing interests.
Open Access
This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,
sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropri-
ate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and
indicate if changes were made. The images or other third party material in this article are included in the
article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is
not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright
holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
1. Bhatt S, Gething PW, Brady OJ, et al. The global distribution and burden of dengue. Nature. 2013;496(7446):504–7. https://
doi.org/10.1038/nature12060.
2. Lindenbach B, Rice C. Flaviviridae: the viruses and their replication. In: Knipe DM, Howley PM, editors. Fields’ Virology. 4th
ed. Lippincott Williams & Wilkins; 2001. p. 991–1041.
3. Vasilakis N, Cardosa J, Diallo M, et al. Sylvatic dengue viruses share the pathogenic potential of urban/endemic dengue viruses.
J Virol. 2010;84(7):3726–8. https://doi.org/10.1128/JVI.02640-09.
4. Fagbami AH, Monath TP, Fabiyi A. Dengue virus infections in Nigeria: a survey for antibodies in monkeys and humans. Trans
R Soc Trop Med Hyg. 1977;71(1):60–5. https://doi.org/10.1016/0035-9203(77)90210-3.
5. Wang E, Ni H, Xu R, et al. Evolutionary Relationships of endemic/epidemic and sylvatic dengue viruses. J Virol.
2000;74(7):3227–34. https://doi.org/10.1128/JVI.74.7.3227-3234.2000.
6. Gubler DJ. Dengue, Urbanization and Globalization: the unholy trinity of the 21st century. Trop Med Health. 2011;39(4
Suppl):3–11. https://doi.org/10.2149/tmh.2011-S05.
46
7. Iwamura T, Guzman-Holst A, Murray KA. Accelerating invasion potential of disease vector Aedes aegypti under climate change.
Nat Commun. 2020;11:2130. https://doi.org/10.1038/s41467-020-16010-4.
8. St. John AL, Rathore APS. Adaptive immune responses to primary and secondary dengue virus infections. Nat Rev Immunol.
2019;19(4):218–30. https://doi.org/10.1038/s41577-019-0123-x.
9. Cattarino L, Rodriguez-Barraquer I, Imai N, Cummings DAT, Ferguson NM. Mapping global variation in dengue transmission
intensity. Sci Transl Med. 2020;12(528):eaax4144. https://doi.org/10.1126/scitranslmed.aax4144.
10. Shepard DS, Undurraga EA, Halasa YA, Stanaway JD. The global economic burden of dengue: a systematic analysis. Lancet
Infect Dis. 2016;16(8):935–41. https://doi.org/10.1016/S1473-3099(16)00146-8.
11. Messina JP, Brady OJ, Golding N, et al. The current and future global distribution and population at risk of dengue. Nat Micro-
biol. 2019;4(9):1508–15. https://doi.org/10.1038/s41564-019-0476-8.
12. Wilder-Smith A, Ooi EE, Horstick O, Wills B. Dengue. The Lancet. 2019;393(10169):350–63. https://doi.org/10.1016/S0140-
6736(18)32560-1.
13. World Health Organization. Dengue guidelines for diagnosis, treatment, prevention and control : new edition. World Health Organi-
zation; 2009. https://apps.who.int/iris/handle/10665/44188. Accessed 19 July 2022.
14. Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century.
Trends Microbiol. 2002;10(2):100–3. https://doi.org/10.1016/S0966-842X(01)02288-0.
15. Beatty ME, Stone A, Fitzsimons DW, et al. Best practices in dengue surveillance: a report from the Asia-Pacific and Americas
dengue prevention boards. Plos Negl Trop Dis. 2010;4(11):e890. https://doi.org/10.1371/journal.pntd.0000890.
16. Syenina A, Jagaraj CJ, Aman SA, Sridharan A, St John AL. St John AL Dengue vascular leakage is augmented by mast cell
degranulation mediated by immunoglobulin Fcγ receptors Medzhitov R, ed. eLife. 2015;4:e05291. https://doi.org/10.7554/
eLife.05291.
17• Rathore APS, Mantri CK, Aman SAB, et al. Dengue virus–elicited tryptase induces endothelial permeability and shock. J Clin
Invest. 2019;129(10):4180–93. https://doi.org/10.1172/JCI128426. This study showed that elevated tryptase levels in dengue
patients correlated with increased severity of DHF and DSS; and tryptase inhibiting drugs may be useful for the treatment of
dengue virus induced vascular leakage and shock.
18. St John AL, Rathore AP, Raghavan B, Ng ML, Abraham SN. Contributions of mast cells and vasoactive products, leukotrienes
and chymase, to dengue virus-induced vascular leakage Medzhitov R, ed. eLife. 2013;2:e00481. https://doi.org/10.7554/eLife.
00481.
19 Rathore AP, Farouk FS, St. John AL. Risk factors and biomarkers of severe dengue. Curr Opin Virol. 2020;43:1–8. https://doi.org/10.1016/j.
coviro.2020.06.008.
20.• Vasikasin V, Rojdumrongrattana T, Chuerboonchai W, et al. Effect of standard dose paracetamol versus placebo as anti-
pyretic therapy on liver injury in adult dengue infection: a multicentre randomised controlled trial. Lancet Glob Health.
2019;7(5):e664–70. https://d oi.o
rg/1
0.1
016/S 2214-1
09X(19)3
0032-4. This clinical trial raises concerns regarding hepatotoxicity
and lack of utility of paracetamol as antipyretic in dengue management.
21. Senaratne T, Carr J, Noordeen F. Elevation in liver enzymes is associated with increased IL-2 and predicts severe outcomes in
clinically apparent dengue virus infection. Cytokine. 2016;83:182–8. https://doi.org/10.1016/j.cyto.2016.04.010.
22. Kittitrakul C, Silachamroon U, Phumratanaprapin W, Krudsood S, Wilairatana P, Treeprasertsuk S. Liver function tests abnor-
mality and clinical severity of dengue infection in adult patients. J Med Assoc Thail Chotmaihet Thangphaet. 2015;98(Suppl
1):S1-8.
23. Trung DT, Thao LTT, Hien TT, et al. Liver involvement associated with dengue infection in adults in Vietnam. Am J Trop Med
Hyg. 2010;83(4):774–80. https://doi.org/10.4269/ajtmh.2010.10-0090.
24. de Souza LJ, Nogueira RMR, Soares LC, et al. The impact of dengue on liver function as evaluated by aminotransferase levels.
Braz J Infect Dis. 2007;11:407–10. https://doi.org/10.1590/S1413-86702007000400007.
25. Deen J, von Seidlein L. Paracetamol for dengue fever: no benefit and potential harm? Lancet Glob Health. 2019;7(5):e552–3.
https://doi.org/10.1016/S2214-109X(19)30157-3.
26. World Health Organization. Regional Office for South-East Asia. Comprehensive Guideline for Prevention and Control of Dengue
and Dengue Haemorrhagic Fever. Revised and Expanded Edition. WHO Regional Office for South-East Asia; 2011. Accessed July
19, 2022. http://apps.who.int/iris/handle/10665/204894
27. Stephen J Thomas, MD, Alan L Rothman, MD, Anon Srikiatkhachorn, MD, Siripen Kalayanarooj, MD. Dengue virus infection:
prevention and treatment - UpToDate. UpToDate. Published February 24, 2022. Accessed June 22, 2022. https://w ww.u ptoda te.
com/contents/dengue-virus-infection-prevention-and-treatment?search=dengue&source=search_result&selectedTitle=3~103&
usage_type=default&display_rank=3#H13085392
28. Wills BA, Dung NM, Loan HT, et al. Comparison of three fluid solutions for resuscitation in dengue shock syndrome. N Engl
J Med. 2005;353(9):877–89. https://doi.org/10.1056/NEJMoa044057.
29. Dung NM, Day NPJ, Tam DTH, et al. Fluid replacement in dengue shock syndrome: a randomized, double-blind comparison
of four intravenous-fluid regimens. Clin Infect Dis. 1999;29(4):787–94. https://doi.org/10.1086/520435.
30. Nhan NT, Phuong CXT, Kneen R, et al. Acute management of dengue shock syndrome: a randomized double-blind comparison
of 4 intravenous fluid regimens in the first hour. Clin Infect Dis. 2001;32(2):204–13. https://doi.org/10.1086/318479.
Dengue: Update on Clinically Relevant Therapeutic Strategies and Vaccines Palanichamy Kala et al. 47
31. Rosenberger KD, Lum L, Alexander N, et al. Vascular leakage in dengue – clinical spectrum and influence of parenteral fluid therapy.
Trop Med Int Health. 2016;21(3):445–53. https://doi.org/10.1111/tmi.12666.
32. Ranjit S, Ramanathan G, Ramakrishnan B, Kissoon N. Targeted interventions in critically ill children with severe dengue. Indian
J Crit Care Med Peer-Rev Off Publ Indian Soc Crit Care Med. 2018;22(3):154–61. https://doi.org/10.4103/ijccm.IJCCM_413_
17.
33. Trung DT, Trieu HT, Wills BA. Microvascular fluid exchange: implications of the revised starling model for resuscitation of dengue shock
syndrome. Front Med. 2020;7:601520. https://doi.org/10.3389/fmed.2020.601520
34.• Masri MFB, Mantri CK, Rathore APS, St John AL. Peripheral serotonin causes dengue virus–induced thrombocytopenia through
5HT2 receptors. Blood. 2019;133(21):2325–37. https://d oi.org/10.1182/blood-2018-08-869156. This preclinical study shows
that drugs targeting serotonin receptor 5HT2 may be useful for the treatment of dengue induced thrombocytopenia.
35. Sun DS, King CC, Huang HS, et al. Antiplatelet autoantibodies elicited by dengue virus non-structural protein 1 cause throm-
bocytopenia and mortality in mice. J Thromb Haemost. 2007;5(11):2291–9. https://d oi.o
rg/1
0.1
111/j.1
538-7
836.2
007.0
2754.x.
36. Ralapanawa U, Alawattegama ATM, Gunrathne M, Tennakoon S, Kularatne SAM, Jayalath T. Value of peripheral blood count
for dengue severity prediction. BMC Res Notes. 2018;11:400. https://doi.org/10.1186/s13104-018-3505-4.
37. Lam PK, Ngoc TV, Thu Thuy TT, et al. The value of daily platelet counts for predicting dengue shock syndrome: results from a
prospective observational study of 2301 Vietnamese children with dengue. Plos Negl Trop Dis. 2017;11(4):e0005498. https://
doi.org/10.1371/journal.pntd.0005498.
38. Lye DC, Archuleta S, Syed-Omar SF, et al. Prophylactic platelet transfusion plus supportive care versus supportive care
alone in adults with dengue and thrombocytopenia: a multicentre, open-label, randomised, superiority trial. The Lancet.
2017;389(10079):1611–8. https://doi.org/10.1016/S0140-6736(17)30269-6.
39.• Chakraborty S, Alam S, Sayem M, et al. Investigation of the efficacy and safety of eltrombopag to correct thrombocytopenia
in moderate to severe dengue patients - a phase II randomized controlled clinical trial. E Clinical Medicine. 2020;29:100624.
https://doi.org/10.1016/j.eclinm.2020.100624. This clinical trial suggests eltrombopag as potential therapeutic to augment
platelet recovery and reduce bleeding manifestations in DHF patients.
40. Jenkins JM, Williams D, Deng Y, et al. Phase 1 clinical study of eltrombopag, an oral, nonpeptide thrombopoietin receptor
agonist. Blood. 2007;109(11):4739–41. https://doi.org/10.1182/blood-2006-11-057968.
41. Assir MZK, Kamran U, Ahmad HI, et al. Effectiveness of platelet transfusion in dengue fever: a randomized controlled trial. Transfus
Med Hemother. 2013;40(5):362–8. https://doi.org/10.1159/000354837.
42. Lee TH, Wong JGX, Leo YS, et al. Potential Harm of Prophylactic Platelet Transfusion in Adult Dengue Patients. PloS Negl Trop
Dis. 2016;10(3):e0004576. https://doi.org/10.1371/journal.pntd.0004576.
43. Ojha A, Nandi D, Batra H, et al. Platelet activation determines the severity of thrombocytopenia in dengue infection. Sci Rep.
2017;7(1):41697. https://doi.org/10.1038/srep41697.
44. Wills BA, Oragui EE, Stephens AC, et al. Coagulation abnormalities in dengue hemorrhagic fever: serial investigations in 167
Vietnamese children with dengue shock syndrome. Clin Infect Dis. 2002;35(3):277–85. https://doi.org/10.1086/341410.
45. Riswari SF, Tunjungputri RN, Kullaya V, et al. Desialylation of platelets induced by Von Willebrand factor is a novel mechanism
of platelet clearance in dengue. Plos Pathog. 2019;15(3):e1007500. https://doi.org/10.1371/journal.ppat.1007500.
46. Tunjungputri RN, Riswari SF, Pramudo SG, et al. Effect of oseltamivir phosphate versus placebo on platelet recovery and plasma
leakage in adults with dengue and thrombocytopenia; a phase 2, multicenter, double-blind, randomized trial. Plos Negl Trop
Dis. 2022;16(1):e0010051. https://doi.org/10.1371/journal.pntd.0010051.
47. Vaughn DW, Green S, Kalayanarooj S, et al. Dengue viremia titer, antibody response pattern, and virus serotype correlate with
disease severity. J Infect Dis. 2000;181(1):2–9. https://doi.org/10.1086/315215.
48. Singla M, Kar M, Sethi T, et al. Immune response to dengue virus infection in pediatric patients in New Delhi, India—asso-
ciation of viremia, inflammatory mediators and monocytes with disease severity. Plos Negl Trop Dis. 2016;10(3):e0004497.
https://doi.org/10.1371/journal.pntd.0004497.
49. Nguyen NM, Tran CNB, Phung LK, et al. A randomized, double-blind placebo controlled trial of balapiravir, a polymerase
inhibitor, in adult dengue patients. J Infect Dis. 2013;207(9):1442–50. https://doi.org/10.1093/infdis/jis470.
50. Tricou V, Minh NN, Van TP, et al. A randomized controlled trial of chloroquine for the treatment of dengue in vietnamese
adults. Plos Negl Trop Dis. 2010;4(8):e785. https://doi.org/10.1371/journal.pntd.0000785.
51. Whitehorn J, Nguyen CVV, Khanh LP, et al. Lovastatin for the treatment of adult patients with dengue: a randomized, double-
blind, placebo-controlled trial. Clin Infect Dis. 2016;62(4):468–76. https://doi.org/10.1093/cid/civ949.
52. Low JG, Sung C, Wijaya L, et al. Efficacy and safety of celgosivir in patients with dengue fever (CELADEN): a phase 1b, ran-
domised, double-blind, placebo-controlled, proof-of-concept trial. Lancet Infect Dis. 2014;14(8):706–15. https://doi.org/10.
1016/S1473-3099(14)70730-3.
53.• Kaptein SJF, Goethals O, Kiemel D, et al. A pan-serotype dengue virus inhibitor targeting the NS3–NS4B interaction. Nature.
2021;598(7881):504–9. https://doi.org/10.1038/s41586-021-03990-6. This study identifies JNJ-A07, a NS4B inhibitor as a
potential direct-acting dengue therapeutic, that showed high efficacy in reducing viremia, viral burden in organs, pro-inflam-
matory cytokine levels and mortality of infected mice.
54. Bardiot D, Koukni M, Smets W, et al. Discovery of indole derivatives as novel and potent dengue virus inhibitors. J Med Chem.
2018;61(18):8390–401. https://doi.org/10.1021/acs.jmedchem.8b00913.
48
55. Zou J, Lee LT, Wang QY, et al. Mapping the interactions between the NS4B and NS3 proteins of dengue virus. J Virol.
2015;89(7):3471–83. https://doi.org/10.1128/JVI.03454-14.
56. Wagstaff KM, Sivakumaran H, Heaton SM, Harrich D, Jans DA. Ivermectin is a specific inhibitor of importin α/β-mediated
nuclear import able to inhibit replication of HIV-1 and dengue virus. Biochem J. 2012;443(Pt 3):851–6. https://doi.org/10.
1042/BJ20120150.
57. Tay MYF, Fraser JE, Chan WKK, et al. Nuclear localization of dengue virus (DENV) 1–4 non-structural protein 5; protection
against all 4 DENV serotypes by the inhibitor Ivermectin. Antiviral Res. 2013;99(3):301–6. https://doi.org/10.1016/j.antiviral.
2013.06.002.
58.• Suputtamongkol Y, Avirutnan P, Mairiang D, et al. Ivermectin Accelerates circulating nonstructural protein 1 (NS1) clearance in adult
dengue patients: a combined phase 2/3 randomized double-blinded placebo controlled trial. Clin Infect Dis. 2021;72(10):e586–93.
https://doi.org/10.1093/cid/ciaa1332. This clinical trial showed Ivermectin reduced NS1 antigen levels in dengue patients, without
any other clinical benefit. Further mechanistic studies are needed to evaluate the utility of Ivermectin as dengue therapeutic.
59. Libraty DH, Young PR, Pickering D, et al. High circulating levels of the dengue virus nonstructural protein ns1 early in dengue illness
correlate with the development of dengue hemorrhagic fever. J Infect Dis. 2002;186(8):1165–8. https://doi.org/10.1086/343813.
60. Avirutnan P, Punyadee N, Noisakran S, et al. Vascular Leakage in severe dengue virus infections: a potential role for the non-
structural viral protein NS1 and complement. J Infect Dis. 2006;193(8):1078–88. https://doi.org/10.1086/500949.
61. Beatty PR, Puerta-Guardo H, Killingbeck SS, Glasner DR, Hopkins K, Harris E. Dengue virus NS1 triggers endothelial perme-
ability and vascular leak that is prevented by NS1 vaccination. Sci Transl Med. 2015;7(304):304ra141-304ra141. https://doi.
org/10.1126/scitranslmed.aaa3787.
62. Puerta-Guardo H, Glasner DR, Harris E. Dengue virus NS1 Disrupts the endothelial glycocalyx, leading to hyperpermeability.
Plos Pathog. 2016;12(7):e1005738. https://doi.org/10.1371/journal.ppat.1005738.
63. Good SS, Shannon A, Lin K, et al. Evaluation of AT-752 a Double prodrug of a guanosine nucleotide analog with in vitro and
in vivo activity against dengue and other flaviviruses. Antimicrob Agents Chemother. 2021;65:e00988.
64. Rothan HA, Mohamed Z, Paydar M, Rahman NA, Yusof R. Inhibitory effect of doxycycline against dengue virus replication
in vitro. Arch Virol. 2014;159(4):711–8. https://doi.org/10.1007/s00705-013-1880-7.
65. Fredeking T, Zavala-Castro J, Gonzalez-Martinez P, et al. Dengue patients treated with doxycycline showed lower mortality
associated to a reduction in IL-6 and TNF levels. Recent Patents Anti-Infect Drug Disc. 2015;10(1):51–8. https://doi.org/10.
2174/1574891X10666150410153839.
66. Pambhar V, Mathur N, Mehta A, et al. Effect of doxycycline and doxycycline with carica papaya on thrombocytopenia and
leucopenia in acute dengue fever patients. J Fam Med Prim Care. 2022;11(6):3270–5. https://doi.org/10.4103/jfmpc.jfmpc_
53_22.
67. Courageot MP, Frenkiel MP, Duarte Dos Santos C, Deubel V, Desprès P. α-Glucosidase inhibitors reduce dengue virus produc-
tion by affecting the initial steps of virion morphogenesis in the endoplasmic reticulum. J Virol. 2000;74(1):564–72.
68. Rathore APS, Paradkar PN, Watanabe S, et al. Celgosivir treatment misfolds dengue virus NS1 protein, induces cellular pro-
survival genes and protects against lethal challenge mouse model. Antiviral Res. 2011;92(3):453–60. https://doi.org/10.1016/j.
antiviral.2011.10.002.
69. Sung C, Wei Y, Watanabe S, et al. Extended Evaluation of virological, immunological and pharmacokinetic endpoints of cel-
aden: a randomized, placebo-controlled trial of celgosivir in dengue fever patients. Plos Negl Trop Dis. 2016;10(8):e0004851.
https://doi.org/10.1371/journal.pntd.0004851.
70. Warfield KL, Plummer EM, Sayce AC, et al. Inhibition of endoplasmic reticulum glucosidases is required for in vitro and in vivo
dengue antiviral activity by the iminosugar UV-4. Antiviral Res. 2016;129:93–8. https://doi.org/10.1016/j.antiviral.2016.03.
001.
71. Perry ST, Buck MD, Plummer EM, et al. An iminosugar with potent inhibition of dengue virus infection in vivo. Antiviral Res.
2013;98(1):35–43. https://doi.org/10.1016/j.antiviral.2013.01.004.
72. Callahan M, Treston AM, Lin G, et al. Randomized single oral dose phase 1 study of safety, tolerability, and pharmacokinetics
of Iminosugar UV-4 Hydrochloride (UV-4B) in healthy subjects. Plos Negl Trop Dis. 2022;16(8):e0010636. https://doi.org/
10.1371/journal.pntd.0010636.
73. Glasner DR, Ratnasiri K, Puerta-Guardo H, Espinosa DA, Beatty PR, Harris E. Dengue virus NS1 cytokine-independent vascular
leak is dependent on endothelial glycocalyx components. Plos Pathog. 2017;13(11):e1006673. https://doi.org/10.1371/journ
al.ppat.1006673.
74. Troost B, Smit JM. Recent advances in antiviral drug development towards dengue virus. Curr Opin Virol. 2020;43:9–21. https://doi.org/10.1016/j.
coviro.2020.07.009.
75 Ah S, Pujar GV, Sethu AK, Bhagyalalitha M, Singh M. Dengue structural proteins as antiviral drug targets: current status in the
drug discovery & development. Eur J Med Chem. 2021;221:113527. https://doi.org/10.1016/j.ejmech.2021.113527.
76. Norshidah H, Vignesh R, Lai NS. Updates on dengue vaccine and antiviral: where are we heading? Molecules. 2021;26(22):6768.
https://doi.org/10.3390/molecules26226768.
77. Qian X, Qi Z. Mosquito-Borne Flaviviruses and Current Therapeutic Advances. Viruses. 2022;14(6):1226. https://doi.org/10.3390/
v14061226.
Dengue: Update on Clinically Relevant Therapeutic Strategies and Vaccines Palanichamy Kala et al. 49
78 Rathore APS, St. john AL. Cross-reactive immunity among flaviviruses. Front Immunol. 2020;11:334. https://doi.org/10.3389/
fimmu.2020.00334.
79 Rey FA, Stiasny K, Vaney MC, Dellarole M, Heinz FX. The bright and the dark side of human antibody responses to flaviviruses:
lessons for vaccine design. EMBO Rep. 2018;19(2):206–24. https://doi.org/10.15252/embr.201745302.
80. Marasco WA, Sui J. The growth and potential of human antiviral monoclonal antibody therapeutics. Nat Biotechnol.
2007;25(12):1421–34. https://doi.org/10.1038/nbt1363.
81. Pierson TC, Xu Q, Nelson S, et al. The stoichiometry of antibody-mediated neutralization and enhancement of West Nile virus
infection. Cell Host Microbe. 2007;1(2):135–45. https://doi.org/10.1016/j.chom.2007.03.002.
82. Magnani DM, Ricciardi MJ, Bailey VK, et al. Dengue virus evades AAV-mediated neutralizing antibody prophylaxis in rhesus
monkeys. Mol Ther. 2017;25(10):2323–31. https://doi.org/10.1016/j.ymthe.2017.06.020.
83. Kam YW, Lee CYP, Teo TH, et al. Cross-reactive dengue human monoclonal antibody prevents severe pathologies and death
from Zika virus infections. JCI Insight. 2017;2(8):e92428. https://doi.org/10.1172/jci.insight.92428.
84. Focosi D, McConnell S, Casadevall A, Cappello E, Valdiserra G, Tuccori M. Monoclonal antibody therapies against SARS-CoV-2.
Lancet Infect Dis. 2022;22(11):e311–26. https://doi.org/10.1016/S1473-3099(22)00311-5.
85. Fibriansah G, Lok SM. The development of therapeutic antibodies against dengue virus. Antiviral Res. 2016;128:7–19.
https://doi.org/10.1016/j.antiviral.2016.01.002.
86. Anasir MI, Poh CL. Discovery of B-cell epitopes for development of dengue vaccines and antibody therapeutics. Med Microbiol
Immunol (Berl). 2022;211(1):1–18. https://doi.org/10.1007/s00430-021-00726-1.
87. Budigi Y, Ong EZ, Robinson LN, et al. Neutralization of antibody-enhanced dengue infection by VIS513, a pan serotype reac-
tive monoclonal antibody targeting domain III of the dengue E protein. Plos Negl Trop Dis. 2018;12(2):e0006209. https://
doi.org/10.1371/journal.pntd.0006209.
88. Ong EZ, Budigi Y, Tan HC, et al. Preclinical evaluation of VIS513, a therapeutic antibody against dengue virus, in non-human
primates. Antiviral Res. 2017;144:44–7. https://doi.org/10.1016/j.antiviral.2017.05.007.
89 Masri MFB, Rathore APS, St John AL. Therapeutics for dengue. Curr Treat Options Infect Dis. 2019;11(3):199–214. https://doi.
org/10.1007/s40506-019-00193-6.
90. Rathore AP, St John AL. Protective and pathogenic roles for mast cells during viral infections. Curr Opin Immunol. 2020;66:74–
81. https://doi.org/10.1016/j.coi.2020.05.003.
91. Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 2010;10(6):440–52. https://doi.
org/10.1038/nri2782.
92. St. John AL, Rathore APS, Yap H, et al. Immune surveillance by mast cells during dengue infection promotes natural killer
(NK) and NKT-cell recruitment and viral clearance. Proc Natl Acad Sci U S A. 2011;108(22):9190–5. https://doi.org/10.1073/
pnas.1105079108.
93. Mantri CK, St. John AL. Immune synapses between mast cells and γδ T cells limit viral infection. J Clin Invest. 2019;129(3):1094–
108. https://doi.org/10.1172/JCI122530.
94. Malavige GN, Wijewickrama A, Fernando S, et al. A preliminary study on efficacy of rupatadine for the treatment of acute
dengue infection. Sci Rep. 2018;8(1):3857. https://doi.org/10.1038/s41598-018-22285-x.
95. Ahmad A, Waseem T, Butt NF, Randhawa FA, Malik U, Shakoori TA. Montelukast Reduces the risk of dengue shock syndrome
in dengue patients. Trop Biomed. 2018;35(4):1115–22.
96.• Sangkaew S, Ming D, Boonyasiri A, et al. Risk predictors of progression to severe disease during the febrile phase of dengue:
a systematic review and meta-analysis. Lancet Infect Dis. 2021;21(7):1014–26. https://doi.org/10.1016/S1473-3099(20)
30601-0. This meta-analysis identifies diabetes as an important risk factor for severe dengue.
97.• Badawi A, Velummailum R, Ryoo SG, et al. Prevalence of chronic comorbidities in dengue fever and West Nile virus: a systematic
review and meta-analysis. Plos One. 2018;13(7):e0200200. https://doi.org/10.1371/journal.pone.0200200. This meta-analysis
identifies obesity, diabetes, and hypertension as the most prevalent comorbidities and risk factors for severe dengue.
98 Tan VPK, Ngim CF, Lee EZ, et al. The association between obesity and dengue virus (DENV) infection in hospitalised patients.
Sekaran SD, ed. Plos One. 2018;13(7):e0200698. https://doi.org/10.1371/journal.pone.0200698.
99. Lee IK, Hsieh CJ, Lee CT, Liu JW. Diabetic patients suffering dengue are at risk for development of dengue shock syndrome/
severe dengue: emphasizing the impacts of co-existing comorbidity(ies) and glycemic control on dengue severity. J Microbiol
Immunol Infect. 2020;53(1):69–78. https://doi.org/10.1016/j.jmii.2017.12.005.
100. Kim J, Yang G, Kim Y, Kim J, Ha J. AMPK activators: mechanisms of action and physiological activities. Exp Mol Med.
2016;48(4):e224–e224. https://doi.org/10.1038/emm.2016.16.
101. Soto-Acosta R, Bautista-Carbajal P, Cervantes-Salazar M, Angel-Ambrocio AH, del Angel RM. DENV up-regulates the
HMG-CoA reductase activity through the impairment of AMPK phosphorylation: a potential antiviral target. Plos Pathog.
2017;13(4):e1006257. https://doi.org/10.1371/journal.ppat.1006257.
102. Jiménez de Oya N, Blázquez AB, Casas J, Saiz JC, Martín-Acebes MA. Direct activation of adenosine monophosphate-activated
protein kinase (AMPK) by PF-06409577 inhibits flavivirus infection through modification of host cell lipid metabolism.
Antimicrob Agents Chemother. 2018;62(7):e00360-18. https://doi.org/10.1128/AAC.00360-18.
50
103.• Htun HL, Yeo TW, Tam CC, Pang J, Leo YS, Lye DC. Metformin use and severe dengue in diabetic adults. Sci Rep. 2018;8(1):3344.
https://doi.org/10.1038/s41598-018-21612-6. This retrospective study showed decreased risk of severe dengue with metformin
use in dengue patients with diabetes.
104 Nguyen NM, Chanh HQ, Tam DTH, et al. Metformin as adjunctive therapy for dengue in overweight and obese patients: a
protocol for an open-label clinical trial (MeDO). Wellcome Open Res. 2020;5:160. https://doi.org/10.12688/wellcomeop
enres.16053.1.
105. Altamish M, Khan M, Baig MS, et al. Therapeutic Potential of medicinal plants against dengue infection: a mechanistic view-
point. ACS Omega. 2022;7(28):24048–65. https://doi.org/10.1021/acsomega.2c00625.
106. Srikanth BK, Reddy L, Biradar S, Shamanna M, Mariguddi DD, Krishnakumar M. An open-label, randomized prospective study to
evaluate the efficacy and safety of Carica papaya leaf extract for thrombocytopenia associated with dengue fever in pediatric subjects.
Pediatr Health Med Ther. 2019;10:5–11. https://doi.org/10.2147/PHMT.S176712.
107. Kasture PN, Nagabhushan KH, Kumar A. A Multi-centric, Double-blind, placebo-controlled, randomized, prospective study
to evaluate the efficacy and safety of Carica papaya leaf extract, as empirical therapy for thrombocytopenia associated with
dengue fever. J Assoc Physicians India. 2016;64(6):15–20.
108. Gadhwal AK, Ankit BS, Chahar C, Tantia P, Sirohi P, Agrawal RP. Effect of Carica papaya leaf extract capsule on platelet count
in patients of dengue fever with thrombocytopenia. J Assoc Physicians India. 2016;64(6):22–6.
109. Nandini C, Madhunapantula SV, Bovilla VR, et al. Platelet enhancement by Carica papaya L. leaf fractions in cyclophospha-
mide induced thrombocytopenic rats is due to elevated expression of CD110 receptor on megakaryocytes. J Ethnopharmacol.
2021;275:114074. https://doi.org/10.1016/j.jep.2021.114074.
110 Erickson KL, Medina EA, Hubbard NE. Micronutrients and innate immunity. J Infect Dis. 2000;182(Supplement_1):S5–10.
https://doi.org/10.1086/315922.
111. Ahmed S, Finkelstein JL, Stewart AM, et al. Micronutrients and dengue. Am J Trop Med Hyg. 2014;91(5):1049–56. https://doi.org/10.4269/
ajtmh.14-0142.
112. Langerman SD, Ververs M. Micronutrient supplementation and clinical outcomes in patients with dengue fever. Am J Trop
Med Hyg. 2021;104(1):45–51. https://doi.org/10.4269/ajtmh.20-0731.
113. Chathurangana PWP, Samaranayake DBDL. Department of Community Medicine, Faculty of Medicine, University of Colombo,
Colombo, Sri Lanka, et al. Effects of vitamin E supplementation on the clinical outcome of dengue fever and dengue haemor-
rhagic fever in children. Asian Pac J Trop Dis. 2017;7(11):645–9. https://doi.org/10.12980/apjtd.7.2017D7-176.
114. Vaish A, Verma S, Agarwal A, Gupta L, Gutch M. Effect of vitamin E on thrombocytopenia in dengue fever. Ann Trop Med
Public Health. 2012;5(4):282. https://doi.org/10.4103/1755-6783.102004.
115. Zaman S, Mahmud MR, Khalid MA, et al. Effectiveness of vitamin d in prevention of dengue haemorrhagic fever and dengue shock
syndrome | Journal of Rawalpindi Medical College. 2017;21:3. http://www.journalrmc.com/index.php/JRMC/article/view/17. Accessed
October 3, 2022.
116. Rerksuppaphol S, Rerksuppaphol L. A Randomized controlled trial of zinc supplementation as adjuvant therapy for dengue
viral infection in Thai children. Int J Prev Med. 2018;9:88. https://doi.org/10.4103/ijpvm.IJPVM_367_17.
117. Ramalingam K, Varghese C, Elias C, Mathew G, Balasubramanian A. A retrospective study on the effect of Vitamin C in the
management of dengue fever in three different states of India. Int J Res Pharm Sci. 2019;10:2670–3. https://doi.org/10.26452/
ijrps.v10i4.1525.
118. Norahmad NA, Mohd Abd Razak MR, Mohmad Misnan N, et al. Effect of freeze-dried Carica papaya leaf juice on inflammatory
cytokines production during dengue virus infection in AG129 mice. BMC Complement Altern Med. 2019;19(1):44. https://
doi.org/10.1186/s12906-019-2438-3.
119. Sharma N, Mishra KP, Chanda S, et al. Evaluation of anti-dengue activity of Carica papaya aqueous leaf extract and its role in
platelet augmentation. Arch Virol. 2019;164(4):1095–110. https://doi.org/10.1007/s00705-019-04179-z.
120. Puerta-Guardo H, De la Cruz Hernández SI, Rosales VH, Ludert JE, del Angel RM. The 1α,25-dihydroxy-vitamin D3 reduces
dengue virus infection in human myelomonocyte (U937) and hepatic (Huh-7) cell lines and cytokine production in the
infected monocytes. Antiviral Res. 2012;94(1):57–61. https://doi.org/10.1016/j.antiviral.2012.02.006.
121. Geneva: World Health Organization; 2013. Guidelines on the quality, safety and efficacy of dengue tetravalent vaccines
(live, attenuated), Annex 2, TRS No 979. Accessed October 5, 2022. http://www.who.int/publications/m/item/TRS_979_
annex-2-dengue
122. Hadinegoro SR, Arredondo-García JL, Capeding MR, et al. Efficacy and long-term safety of a dengue vaccine in regions of
endemic disease. N Engl J Med. 2015;373(13):1195–206. https://doi.org/10.1056/NEJMoa1506223.
123• Rivera L, Biswal S, Sáez-Llorens X, et al. Three-year efficacy and safety of Takeda’s dengue vaccine candidate (TAK-003). Clin
Infect Dis Off Publ Infect Dis Soc Am. 2021;75(1):107–17. https://doi.org/10.1093/cid/ciab864. Latest efficacy results (36
months) from the TIDES trial that allowed for approval of Takeda’s Qdenga vaccine to be used in Indonesia and marketing
approval in the European Union.
124. Kallas EG, Precioso AR, Palacios R, et al. Safety and immunogenicity of the tetravalent, live-attenuated dengue vaccine
Butantan-DV in adults in Brazil: a two-step, double-blind, randomised placebo-controlled phase 2 trial. Lancet Infect Dis.
2020;20(7):839–50. https://doi.org/10.1016/S1473-3099(20)30023-2.
Dengue: Update on Clinically Relevant Therapeutic Strategies and Vaccines Palanichamy Kala et al. 51
125. Diaz C, Koren M, Lin L, et al. Safety and Immunogenicity of different formulations of a tetravalent dengue purified inactivated
vaccine in healthy adults from Puerto Rico: Final Results after 3 Years of Follow-Up from a Randomized, Placebo-Controlled
Phase I Study. Am J Trop Med Hyg. 2020;102(5):951–4. https://doi.org/10.4269/ajtmh.19-0461.
126. Bauer K, Esquilin IO, Cornier AS, et al. A Phase II, Randomized, 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(3):441–53. https://
doi.org/10.4269/ajtmh.14-0625.
127. Beckett CG, Tjaden J, Burgess T, et al. Evaluation of a prototype dengue-1 DNA vaccine in a phase 1 clinical trial.
Vaccine. 2011;29(5):960–8. https://d oi.o rg/10.1016/j.v accin e.2 010.11.0 50.
128. Danko JR, Kochel T, Teneza-Mora N, et al. Safety and immunogenicity of a tetravalent dengue dna vaccine administered with a cationic lipid-
based adjuvant in a phase 1 clinical trial. Am J Trop Med Hyg. 2018;98(3):849–56. https://doi.org/10.4269/ajtmh.17-0416.
129. Manoff SB, Sausser M, Falk Russell A, et al. Immunogenicity and safety of an investigational tetravalent recombinant subu-
nit vaccine for dengue: results of a Phase I randomized clinical trial in flavivirus-naïve adults. Hum Vaccines Immunother.
2019;15(9):2195–204. https://doi.org/10.1080/21645515.2018.1546523.
130. Capeding MR, Tran NH, Hadinegoro SRS, et al. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy
children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. The Lancet. 2014;384(9951):1358–65.
https://doi.org/10.1016/S0140-6736(14)61060-6.
131. Villar L, Dayan GH, Arredondo-García JL, et al. Efficacy of a tetravalent dengue vaccine in children in Latin America. N Engl J
Med. 2015;372(2):113–23. https://doi.org/10.1056/NEJMoa1411037.
132. Sabchareon A, Wallace D, Sirivichayakul C, et al. Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue
vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. The Lancet. 2012;380(9853):1559–67. https://doi.
org/10.1016/S0140-6736(12)61428-7.
133. Halstead SB. Dengvaxia sensitizes seronegatives to vaccine enhanced disease regardless of age. Vaccine. 2017;35(47):6355–8.
https://doi.org/10.1016/j.vaccine.2017.09.089.
134. Sridhar S, Luedtke A, Langevin E, et al. Effect of dengue serostatus on dengue vaccine safety and efficacy. N Engl J Med.
2018;379(4):327–40. https://doi.org/10.1056/NEJMoa1800820.
135. World Health Organization - Weekly epidemiological report. Dengue vaccines: WHO position paper. 2018;93:457–476. http://
www.who.int/publications-detail-redirect/who-wer9335-457-476 Revised recommendations for immunization with Deng-
vaxia from WHO Strategic Advisory Group of Experts on immunization committee based on evidence indicating increased
hospitalisation and severe dengue risk in seronegative individuals when administered the vaccine.
136. Badan Pengawas Obat dan Makanan - Republik Indonesia. Approval of Dengue Vaccine Distribution Permit (Qdenga) for
Age 6–45 Years. BPOM Press Release. Published September 8, 2022. https://www.pom.go.id/new/view/more/pers/659/Perse
tujuan-Izin-Edar-Vaksin-Dengue--Qdenga--untuk-Usia-6---45-Tahun.html. Accessed 3 April 2023.
137. European Medicines Agency. Qdenga. European Medicines Agency. Published October 12, 2022. https://www.ema.europa.eu/
en/medicines/human/EPAR/qdenga. Accessed 2 April 2023.
138. Takeda. Takeda’s QDENGA®▼ (Dengue Tetravalent Vaccine [Live, Attenuated]) Approved for Use in European Union (Takeda
Newsroom, 2022). https://w ww.takeda.com/newsroom/newsreleases/2022/takedas-qdenga-dengue-tetravalent-vaccine-live-
attenuated-approved-for-use-in-european-union. Accessed December 11, 2022.
139. Takeda. Takeda’s QDENGA (Dengue Tetravalent Vaccine [Live, Attenuated]) Approved in Indonesia for Use Regardless
of Prior Dengue Exposure. (Takeda Newsroom, 2022). https://w ww.t akeda.c om/n ewsro om/n ewsre lease s/2022/t aked
as-qdenga-dengue-tetravalent-vaccine-live-attenuated-approved-in-indonesia-for-use-regardless-of-prior-dengue-expos
ure/. Accessed 10 October 2022.
140. Huang CYH, Butrapet S, Tsuchiya KR, Bhamarapravati N, Gubler DJ, Kinney RM. Dengue 2 PDK-53 virus as a chimeric carrier
for tetravalent dengue vaccine development. J Virol. 2003;77(21):11436–47. https://doi.org/10.1128/JVI.77.21.11436-11447.
2003.
141. Biswal S, Reynales H, Saez-Llorens X, et al. Efficacy of a tetravalent dengue vaccine in healthy children and adolescents. N
Engl J Med. 2019;381(21):2009–19. https://doi.org/10.1056/NEJMoa1903869.
142. Biswal S, Borja-Tabora C, Martinez Vargas L, et al. Efficacy of a tetravalent dengue vaccine in healthy children aged 4–16 years: a
randomised, placebo-controlled, phase 3 trial. The Lancet. 2020;395(10234):1423–33. https://d oi.o
rg/1
0.1
016/S 0140-6 736(20)
30414-1.
143. López-Medina E, Biswal S, Saez-Llorens X, et al. Efficacy of a dengue vaccine candidate (TAK-003) in healthy children and
adolescents 2 years after vaccination. J Infect Dis. 2022;225(9):1521–32. https://doi.org/10.1093/infdis/jiaa761.
144. de Silva A. Safety of dengue vaccine? Clin Infect Dis. 2023;76(2):371–2. https://doi.org/10.1093/cid/ciac690.
145. Tricou V, Gottardo R, Egan MA, et al. Characterization of the cell-mediated immune response to Takeda’s live-attenuated
tetravalent dengue vaccine in adolescents participating in a phase 2 randomized controlled trial conducted in a dengue-
endemic setting. Vaccine. 2022;40(8):1143–51. https://doi.org/10.1016/j.vaccine.2022.01.016.
146. Whitehead SS. Development of TV003/TV005, a single dose, highly immunogenic live attenuated dengue vaccine; what makes
this vaccine different from the Sanofi-Pasteur CYD™ vaccine? Expert Rev Vaccines. 2016;15(4):509–17. https://d oi.o rg/1
0.1
586/
14760584.2016.1115727.
52
147. Durbin AP, Kirkpatrick BD, Pierce KK, et al. A single dose of any of four different live attenuated tetravalent dengue vaccines is
safe and immunogenic in flavivirus-naive adults: a randomized, double-blind clinical trial. J Infect Dis. 2013;207(6):957–65.
https://doi.org/10.1093/infdis/jis936.
148. Kirkpatrick BD, Durbin AP, Pierce KK, et al. Robust and balanced immune responses to all 4 dengue virus serotypes following
administration of a single dose of a live attenuated tetravalent dengue vaccine to healthy, flavivirus-naive adults. J Infect Dis.
2015;212(5):702–10. https://doi.org/10.1093/infdis/jiv082.
149. Durbin AP. Historical discourse on the development of the live attenuated tetravalent dengue vaccine candidate TV003/TV005.
Curr Opin Virol. 2020;43:79–87. https://doi.org/10.1016/j.coviro.2020.09.005.
150. Kirkpatrick BD, Whitehead SS, Pierce KK, et al. The live attenuated dengue vaccine TV003 elicits complete protection against
dengue in a human challenge model. Sci Transl Med. 2016;8(330):330ra36-330ra36 (10.1126/scitranslmed.aaf1517).