EMERGING INTERVENTIONS & THERAPIES FOR EFFECTIVE DENGUE MANAGEMENT
HTML Full TextEMERGING INTERVENTIONS & THERAPIES FOR EFFECTIVE DENGUE MANAGEMENT
Gourav Ganguly *, Tarpan Sahoo, Shibam Ghosh, Abhik Gupta, Arabinda Nayak, S. K. Mojamel and Mirza Sahabuddin
Department of Pharmacology, Bengal College of Pharmaceutical Technology, Dubrajpur, Birbhum, West Bengal, India.
ABSTRACT: The review delves into the latest developments in dengue fever treatment, particularly focusing on emerging therapies such as vaccines and host-directed therapies (HDTs) designed to bolster the immune response to the virus. It outlines the progress in vaccine research, with a particular emphasis on the introduction of Dengvaxia, the first licensed dengue vaccine, and highlights ongoing efforts to develop more effective vaccines that offer broader protection across different serotypes of the virus. The review also underscores the importance of a holistic approach to managing dengue fever. It argues that alongside medical interventions, factors such as dietary considerations and community engagement are crucial in prevention strategies. It stresses the need for integrated public health initiatives that empower local communities to take proactive measures in controlling mosquito populations and reducing the risk of transmission. Furthermore, the review calls for sustained research efforts and collaboration across multiple sectors, including healthcare professionals, scientists, and policymakers. By fostering international cooperation, the hope is to develop innovative solutions that can effectively reduce the global burden of dengue fever. The ultimate goal is not only to advance vaccine development but also to implement comprehensive strategies that combine prevention, treatment, and community involvement, ensuring that global health initiatives are robust and sustainable in the fight against dengue fever.
Keywords: Approach, Broader, Communities, Dengue, Engagement
INTRODUCTION: Dengue viruses have rapidly expanded within countries and across regions over the past few decades, leading to an increase in the frequency of epidemics and severe dengue cases, as well as the hyperendemicity of multiple dengue virus serotypes in many tropical nations. Additionally, there has been local transmission in parts of Indie 1.
Today, dengue is recognized as the most widespread and swiftly spreading mosquito-borne viral disease affecting humans. Notably, the last decade has witnessed a significant rise in research focused on dengue virology, pathogenesis, and immunology, along with advancements in the development of antivirals, vaccines, and innovative vector-control strategies, all of which could greatly enhance dengue prevention and control efforts 2.
The term "dengue fever" became widely used only after 1828. Dengue viruses (DV) belong to the Flaviviridae family and consist of four serotypes known as DV-1, DV-2, DV-3, and DV-4. DV is a positive-stranded RNA virus encased in a protein shell, containing three structural protein genes that code for the nucleocapsid (C) protein, a membrane-associated (M) protein, and an envelope (E) glycoprotein, along with seven non-structural (NS) proteins. The primary vectors for transmission are the Aedes aegypti mosquito and, to a lesser extent, Aedes albopictus. All four serotypes can cause a range of illnesses, from asymptomatic infections to mild, self-limiting dengue fever (DF) and severe forms that can be fatal, such as dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS) 3.
The World Health Organization's 2009 classification divides dengue fever into two categories: uncomplicated and severe, although the older 1997 classification is still commonly referenced. The 1997 classification includes undifferentiated fever, dengue fever (DF), and dengue hemorrhagic fever (DHF). Four key manifestations of dengue illness are: (i) a persistent high fever lasting 2 to 7 days; (ii) a tendency for bleeding, indicated by a positive tourniquet test, petechiae, or nosebleeds; (iii) thrombocytopenia (platelet count below 100×10^9/l); and (iv) signs of plasma leakage, which may be reflected in hemoconcentration (an increase in hematocrit of 20% or more above the average for the individual’s age, sex, and population), pleural effusion, and ascites. Significant research has been conducted at various centers in India on the molecular epidemiology of dengue, its immunopathology, and vaccine development 4-5.
The global impact of dengue is significant, with an estimated 50 million infections occurring annually across about 100 countries, and the potential for further spread. A key factor in the rise of dengue as a public health concern has been the distribution of effective mosquito vectors in many tropical and subtropical regions. The primary vector, the urban-adapted Aedes aegypti mosquito, has become prevalent in these areas. This mosquito originated in Africa during the slave trade between the 15th and 19th centuries, later spreading to Asia through commercial trade in the 18th and 19th centuries, and it has continued to expand globally due to increased travel and trade over the past 50 years 6.
Additionally, the geographic range of a secondary vector, Aedes albopictus, has rapidly increased in recent years. The globalization of trade, particularly in used tires, is believed to have facilitated the movement of eggs and immature forms of these virus-carrying mosquitoes into new regions. The rise of dengue has also been supported by rapid urbanization in Asia and Latin America, which has led to higher population densities and a greater number of breeding sites for vectors in densely populated urban areas and their surroundings. While dengue infections in Africa are largely unmeasured, recent outbreaks indicate that many regions of the continent may be at risk for heightened transmission. More surveillance is necessary to accurately assess the true burden of the disease 7.
Vector control remains the primary strategy for dengue prevention, focusing on chemically or biologically targeting mosquitoes and eliminating their breeding sites. Unfortunately, this approach has largely failed to prevent disease transmission in nearly all countries where dengue is endemic. A significant factor in this challenge is the antigenic diversity of the dengue virus; the lack of long-term cross-immunity among its four serotypes allows individuals to experience multiple infections over time 8.
The ongoing spread of dengue highlights how global trade (along with the movement of mosquito vectors), increased travel within and between countries (including the transport of infected individuals), urban overcrowding (which promotes multiple infections from a single mosquito), and ineffective vector control strategies have contributed to the pandemic. As dengue continues to spread globally, healthcare professionals in temperate regions such as North America, Europe, Australia, and Japan are more likely than ever to encounter travelers returning with dengue infections. Therefore, clinicians should consider dengue as a possible diagnosis for any patient presenting with fever that arises within 14 days of even a brief visit to tropical or subtropical areas, including places where dengue has not traditionally been viewed as endemic 9, 10.
Etiology: Dengue infection is present in the blood of infected individuals. When an Aedes mosquito bites a person with dengue fever, it ingests blood that contains the dengue virus. The virus then undergoes further development inside the mosquito’s body over several days. If this infected mosquito subsequently bites a healthy person, the virus is transmitted into their bloodstream, leading to infection and the potential onset of dengue fever symptoms 11.
Transmission through Mosquito Bite: The infection is transmitted to humans through the bites of infected female mosquitoes, primarily the Aedes aegypti species. Other species within the Aedes genus can also act as vectors, but their role is secondary to Aedes aegypti. After feeding on a dengue virus-infected person, the virus replicates in the mosquito's midgut before spreading to other tissues, including the salivary glands. The time from when the mosquito ingests the virus to when it can transmit it to another host is known as the extrinsic incubation period (EIP). This period typically lasts about 8 to 12 days when temperatures are between 25-28°C. Variations in the EIP are influenced not only by temperature but also by factors such as daily temperature fluctuations, the genotype of the virus, and the initial viral load. Once a mosquito becomes infectious, it can transmit the virus for the remainder of its life 12.
FIG. 1: LIFE CYCLE OF DENGUE
Human-to-mosquito Transmission: Mosquitoes can become infected with the dengue virus (DENV) from individuals who are viremic, meaning they have the virus in their blood. This includes people with symptomatic dengue infections, those who are pre-symptomatic and even individuals who show no symptoms at all (asymptomatic). Transmission from humans to mosquitoes can occur as early as two days before symptoms appear and up to two days after the fever has resolved. The likelihood of mosquitoes becoming infected is positively associated with high levels of viremia and fever in the patient. Conversely, high levels of DENV-specific antibodies in the patient are linked to a reduced risk of mosquito infection (Nguyen et al., 2013, PNAS). Most people remain viremic for about 4 to 5 days 13.
Maternal Transmission: The primary mode of dengue virus (DENV) transmission among people involves mosquito vectors. However, there is also evidence suggesting the possibility of maternal transmission, where the virus can be passed from a pregnant mother to her child. Although the rates of vertical transmission seem to be low, the risk appears to be associated with the timing of the dengue infection during pregnancy. When a mother contracts DENV while pregnant, the child may face complications such as preterm birth, low birth weight, and fetal distress 14.
Other Transmission Modes: There have been notable cases of dengue virus transmission through blood products, organ donations, and breastfeeding. Additionally, transovarial transmission of the virus has also been observed in mosquitoes.
Current Situation: Dengue is endemic to tropical and subtropical countries and is the fastest-spreading arboviral disease in these regions. It can also behave in an epidemic manner when conditions are favorable. Key factors that promote both endemic and epidemic outbreaks include the presence of extensive areas infested with Aedes mosquitoes, significant populations of susceptible humans, and the ongoing introduction and/or circulation of one or more serotypes of the virus 15.
Epidemiology:
Disease Burden and Epidemiology: Dengue is ranked as the second most serious vector-borne disease globally, following malaria, in terms of incidence and mortality rates. In recent years, the economic impact of dengue has risen significantly in India. In several countries, including Bhutan, Brunei, Cambodia, East Timor, Indonesia, Myanmar, the Philippines, Singapore, Vietnam, and Taiwan, the annual economic burden has reached approximately $950 million. The disease burden of dengue is estimated at 25.5 disability-adjusted life years per 100,000 people. However, the true incidence and economic impact of dengue are likely underestimated due to many cases being asymptomatic and underreported 16.
FIG. 2: GEOGRAPHICAL DISTRIBUTION
Geographic Distribution: Dengue is endemic in over 100 countries and regions in tropical and subtropical areas, including Southeast Asia, Central and South America, Africa, the Western Pacific, and the Eastern Mediterranean. According to WHO estimates, around 2.5 billion people live in these endemic regions, with 50 million cases of dengue virus infection occurring each year, resulting in approximately 25,000 fatalities.
Notably, the incidence rate has surged 30-fold over the past 50 years, with a rapid geographical expansion. Furthermore, the incidence in various countries has increased fivefold in the last decade. A 2013 study indicated that approximately 3.6 billion people worldwide reside in areas at risk for dengue, with around 390 million infections each year and 96 million symptomatic cases. Of these, about 500,000 individuals develop severe dengue, such as dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). Recently, dengue morbidity has risen sharply, with significant outbreaks occurring in Southeast Asia and China in 2014. Cases of dengue have also been reported in higher latitude regions, including Florida (USA), Southeastern France, Madeira Island, Spain, Croatia, and Japan. In Southern China, the frequency and scale of dengue transmission have been increasing over recent decades, along with a rise in geographical spread, incidence, and severe dengue cases, which is worsening currently 17-18.
Population Distribution: Anyone can be infected with the dengue virus (DENV), with approximately 30% to 60% exhibiting symptoms. However, there are notable differences between endemic and non-endemic areas. In non-endemic regions, young adults have a higher infection rate, while in endemic areas, children experience the highest incidence, accounting for 85% of total cases. There are no significant differences in infection rates based on gender or occupation, although the shift toward higher rates in adults requires further investigation. The likelihood of infection is closely linked to the probability of being bitten by infected Aedes mosquitoes. Medical workers are considered high-risk due to inadequate infection control measures, leading to medical facilities becoming outbreak hotspots. This results in the virus being transmitted from infected Aedes mosquitoes to other patients, healthcare workers, and visitors, thereby exacerbating the outbreak 19.
TABLE 1: DENGUE CASES AND DEATHS IN INDIA SINCE 2019
Sl. no. | Affected States/UTs | 2019 | 2020 | 2021 | 2022 | 2023 | 2024* | ||||||
C | D | C | D | C | D | C | D | C | D | C | D | ||
1 | Andhra Pradesh | 5286 | 0 | 925 | 0 | 4760 | 0 | 6391 | 0 | 6453 | 0 | 4790 | 0 |
2 | Arunachal Pradesh | 123 | 0 | 1 | 0 | 7 | 0 | 114 | 0 | 130 | 0 | 18 | 0 |
3 | Assam | 196 | 0 | 33 | 0 | 103 | 0 | 1826 | 2 | 8208 | 7 | 1553 | 0 |
4 | Bihar | 6712 | 0 | 493 | 2 | 633 | 2 | 13972 | 32 | 20224 | 74 | 7338 | 15 |
5 | Chattisgarh | 722 | 0 | 57 | 0 | 1086 | 0 | 2679 | 10 | 2412 | 0 | 3261 | 0 |
6 | Goa | 992 | 0 | 376 | 0 | 649 | 0 | 443 | 1 | 512 | 3 | 512 | 0 |
7 | Gujarat | 18219 | 17 | 1564 | 2 | 10983 | 14 | 6682 | 7 | 7222 | 7 | 6008 | 5 |
8 | Haryana | 1207 | 0 | 1377 | 0 | 11835 | 13 | 8996 | 18 | 8081 | 11 | 3789 | 3 |
9 | Himachal Pradesh | 344 | 2 | 21 | 0 | 349 | 0 | 3326 | 1 | 1989 | 0 | 3178 | 0 |
10 | J & K | 439 | 0 | 53 | 0 | 1709 | 4 | 8269 | 18 | 6403 | 10 | 4549 | 1 |
11 | Jharkhand | 825 | 0 | 79 | 0 | 220 | 1 | 290 | 0 | 2578 | 4 | 1172 | 1 |
12 | Karnataka | 16986 | 13 | 3823 | 0 | 7393 | 7 | 9889 | 9 | 19300 | 11 | 30973 | 16 |
13 | Kerala | 4652 | 16 | 4399 | 5 | 3251 | 27 | 4432 | 29 | 17426 | 153 | 18534 | 71 |
14 | Lakshadweep | 0 | 0 | 0 | 0 | 1 | 0 | 67 | 0 | 445 | 0 | 469 | 0 |
15 | Madhya Pradesh | 4189 | 2 | 806 | 0 | 15592 | 11 | 3318 | 2 | 6979 | 0 | 7941 | 1 |
16 | Meghalaya | 82 | 0 | 4 | 0 | 129 | 0 | 26 | 0 | 114 | 0 | 63 | 0 |
17 | Maharashtra | 14907 | 29 | 3356 | 10 | 12720 | 42 | 8578 | 27 | 19034 | 55 | 16845 | 26 |
18 | Manipur | 359 | 0 | 37 | 0 | 203 | 0 | 503 | 4 | 2548 | 0 | 2022 | 5 |
19 | Mizoram | 42 | 0 | 67 | 0 | 83 | 0 | 1868 | 5 | 2060 | 2 | 528 | 1 |
20 | Nagaland | 8 | 0 | 1 | 0 | 24 | 0 | 154 | 0 | 4943 | 2 | 24 | 0 |
21 | Odisha | 3758 | 4 | 496 | 0 | 7548 | 0 | 7063 | 0 | 12845 | 1 | 8771 | 0 |
22 | Punjab | 10289 | 14 | 8435 | 22 | 23389 | 55 | 11030 | 41 | 13687 | 39 | 2634 | 0 |
23 | Rajasthan | 13706 | 17 | 2023 | 7 | 20749 | 96 | 13491 | 10 | 13924 | 14 | 10458 | 3 |
24 | Sikkim | 444 | 0 | 11 | 0 | 243 | 1 | 264 | 0 | 311 | 0 | 231 | 0 |
25 | Tamil Nadu | 8527 | 5 | 2410 | 0 | 6039 | 8 | 6430 | 8 | 9121 | 12 | 19138 | 7 |
26 | Tripura | 114 | 0 | 24 | 0 | 349 | 0 | 56 | 0 | 1447 | 0 | 774 | 0 |
27 | Telangana | 13331 | 7 | 2173 | 0 | 7135 | 0 | 8972 | 0 | 8016 | 1 | 9761 | 0 |
28 | Uttar Pradesh | 10557 | 26 | 3715 | 6 | 29750 | 29 | 19821 | 33 | 35402 | 36 | 10234 | 2 |
29 | Uttrakhand | 10622 | 8 | 76 | 1 | 738 | 2 | 2337 | 0 | 4320 | 17 | 400 | 0 |
30 | West Bengal* | NR | NR | 5166 | 0 | 8264 | 7 | 67271 | 30 | 30683 | 4 | 441 | 0 |
31 | A& N Island | 168 | 0 | 98 | 0 | 175 | 0 | 1014 | 3 | 846 | 0 | 58 | 0 |
32 | Chandigarh | 286 | 0 | 265 | 0 | 1596 | 3 | 910 | 1 | 454 | 0 | 199 | 0 |
33 | Delhi | 5077 | 0 | 1269 | 0 | 13089 | 23 | 10183 | 9 | 16866 | 19 | 5637 | 3 |
34 | D&N Haveli | 1491 | 2 | 248 | 0 | 547 | 0 | 685 | 0 | 1178 | 0 | 333 | 0 |
35 | Daman & Diu | 625 | 2 | 71 | 0 | 279 | 0 | 228 | 0 | 284 | 1 | 242 | 0 |
36 | Puduchery | 2030 | 2 | 633 | 1 | 1625 | 1 | 1673 | 3 | 2790 | 2 | 3689 | 0 |
Total | 157315 | 166 | 44585 | 56 | 193245 | 346 | 233251 | 303 | 289235 | 485 | 186567 | 160 |
Risk Factors for Transmission:
Source of Infection: Humans serve as the primary amplifying hosts for dengue virus (DENV), with cases acting as the main source of infection. The communicable period ranges from one day before to five days after the onset of symptoms, during which Aedes mosquitoes can transmit the virus to healthy individuals. Notably, over 50% of infected individuals may experience asymptomatic infections, with a ratio of asymptomatic to symptomatic infections of 2.2:1. These asymptomatic cases significantly contribute to the virus's transmission. Non-human primates, including chimpanzees, gibbons, and macaques, are important natural reservoirs, particularly in jungle epidemic regions of Southeast Asia. Other animals, such as monkeys, bats, pigs, and chickens, also serve as reservoirs and transmitters of the virus. In jungle environments, Aedes mosquitoes residing in the tree canopy can spread the virus among primates and subsequently infect humans, leading to outbreaks in densely populated villages and cities, and even globally via shipping and air travel. In urban and rural settings, the transmission pattern follows an Aedes-human-Aedes cycle, as observed in regions like Guangdong, Hainan, Guangxi, Fujian, and Zhejiang in China. While vertical transmission of DENV in mosquito vectors has been confirmed in laboratory studies, it has not been thoroughly documented in natural environments 20-21.
Route of Transmission: Aedes mosquitoes are the primary vectors for transmitting dengue virus (DENV), with around 13 subspecies capable of this transmission. Among them, Aedes aegypti, commonly known as the yellow fever mosquito, predominantly inhabits tropical regions and is the most significant and frequent vector for biting humans, especially before it reproduces, as it tends to live near human populations. Aedes albopictus, or the Asian tiger mosquito, has a broader geographic distribution, being found not only in Asia and other endemic areas but also in Europe and North America, where it has been responsible for significant outbreaks in recent years.
The role of Culex fatigans as a potential reservoir is still uncertain. Additionally, dengue can be transmitted through blood products and organ transplantation, with infection rates in Singapore reported to be as high as 1.6%–6.0%. Other transmission routes in humans are considered rare 22.
Susceptible Population and Mechanism: In regions where dengue virus (DENV) transmission is new or has been introduced, populations are largely vulnerable to infection, with adults experiencing a higher incidence. Conversely, in endemic areas, most adults possess specific antibodies, leading to a higher incidence of infections among children. This pattern arises from the build-up of immunity in the adult population over time. After a DENV infection, individuals generate serotype-specific antibodies that can last for several years or even a lifetime. However, immunity against different serotypes is temporary, lasting anywhere from 2 months to 2 years. Notably, secondary infections with different serotypes can increase the risk of severe dengue manifestations like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Infants with maternal antibodies are particularly at risk for DHF/DSS if they become infected.
The phenomenon known as antibody-dependent enhancement (ADE) suggests that antibodies from a previous infection can worsen disease severity upon reinfection. Factors such as the type of virus and the time between infections can influence the severity of the secondary infection, with longer intervals typically associated with worse outcomes. The impact of heterotypic antibodies on overall population immunity has not been extensively studied. Research conducted in Nicaragua and Peru has shown instances of reinfection with the same serotype 23. Additionally, individuals with pre-existing health conditions, such as diabetes, asthma, G6PD deficiency, and sickle-cell anemia, are known to be at an increased risk for severe dengue. Dengue virus (DENV), a member of the Flaviviridae family and the Flavivirus genus, is an enveloped icosahedral virus with a diameter of 40–50 nm. It comprises four distinct serotypes: DENV-1, DENV-2, DENV-3, and DENV-4, which share approximately 60%–70% similarity in their amino acid sequences. The DENV genome is a single positive RNA strand, about 11 kb long, featuring a single open reading frame that accounts for 96% of its nucleotides encoding viral proteins 24. This genome is organized into two parts: the 5' end (one-quarter) encodes three structural proteins capsid (C), membrane (M), and envelope (E) while the 3' end (three-quarters) encodes seven non-structural (NS) proteins: NS1, NS2a, NS2b, NS3, NS4a, NS4b, and NS5. In 2013, DENV-5 was identified in Malaysia, stemming from an infection that occurred in 2007, suggesting it had been present in primate populations in forests for an extended period, complicating vaccine development efforts. Phylogenetic studies indicate that DENV diversity has been long-standing, with lineage replacements leading to fluctuations in diversity levels. Since World War II, the four serotypes have exhibited rapid diversity, resulting in new genotypic variations. The nucleotide sequence differences between serotypes are significant, around 35%, while variations within the same serotype, which can have multiple genotypes, show approximately 3% differences at the amino acid level and 6% at the nucleotide level. These differences in serotype, genotype, and clade can lead to variations in clinical manifestations, disease severity, virulence, and epidemic potential 25.
Control and Prevention Strategy: The overarching aim of dengue control is to swiftly identify cases, prevent secondary infections, avert major outbreaks, and mitigate the risks associated with dengue. While vaccines may contribute to prevention, the primary focus remains on vector control to interrupt transmission routes. A crucial aspect of this strategy is environmental management and the elimination of mosquito breeding sites. Dengue is recognized as an environmental and community disease, often described as a behavioral and ecological issue. Effective disease control requires the collaboration of multiple departments rather than placing the burden on a single individual, organization, or governmental system. Moreover, relying on isolated measures is insufficient to manage dengue effectively. The WHO strategy aimed to reduce mortality and incidence rates by 50% and 25%, respectively, by 2020, through four key approaches: advanced vector control technologies, improved diagnostic methods, evidence-based clinical interventions, and candidate vaccines. In mainland China, the focus is on enhancing surveillance, controlling vector populations, preventing imported cases, improving diagnosis and treatment, and fostering multidisciplinary collaboration and community engagement 26.
Pathophysiology of Dengue: Aedes aegypti is the main mosquito vector for dengue fever viruses and has a strong association with humans and their living environments. Additionally, the dengue virus can suppress the human immune response.
The Vector is the Aedes aegypti Mosquito:
- Aedes aegypti, the main mosquito vector for dengue viruses, is an insect that closely interacts with humans and their living spaces. Humans provide both blood meals and water-filled containers necessary for the mosquitoes’ development.
- The female mosquito lays her eggs on the walls of these containers, and the eggs hatch into larvae after rain or flooding occurs.
- The larvae transform into pupae in about a week and then into adult mosquitoes within two days 27.
The Pathogen is Dengue Fever Virus: Dengue fever virus (DENV) is an RNA virus belonging to the Flaviviridae family and the Flavivirus genus. Other viruses in this genus include the yellow fever virus, West Nile virus, St. Louis encephalitis virus, Japanese encephalitis virus, tick-borne encephalitis, Kyasanur Forest disease virus, and Omsk hemorrhagic fever virus. Most of these viruses are transmitted by arthropods, such as mosquitoes and ticks, and are commonly known as arboviruses (arthropod-borne viruses).
The DENV genome comprises approximately 11,000 nucleotide bases that encode three structural proteins (C, prM, and E) essential for forming the virus particle, as well as seven non-structural proteins (NS1, NS2a, NS2b, NS3, NS4a, NS4b, and NS5) that are found only in infected host cells and are crucial for the virus's replication. There are five strains of the virus, with the first four identified as DENV-1, DENV-2, DENV-3, and DENV-4. The differences between these serotypes are based on their antigenic properties 28.
Transmission: Dengue virus is mainly spread by Aedes mosquitoes, especially A. aegypti, which typically bite during the day, particularly in the early morning and evening. However, they can bite and transmit the virus at any time throughout the year. Other Aedes species that also spread the disease include A. albopictus, A. polynesiensis, and A. scutellaris. While humans are the primary hosts for the virus, it can also circulate in nonhuman primates. A single bite from an infected mosquito can lead to infection. When a female mosquito takes a blood meal from a dengue-infected person during the first 2 to 10 days of their fever, the mosquito becomes infected as the virus enters the cells lining its gut. After about 8 to 10 days, the virus spreads to other tissues, including the salivary glands, and is then released into the mosquito's saliva. The virus does not appear to harm the mosquito, which remains infected for life. Aedes aegypti prefers to lay its eggs in artificial water containers, live near humans, and feed primarily on people. Dengue can also be transmitted through infected blood products, organ donations, and vertical transmission from mother to child. Although there have been reports of other rare modes of person-to-person transmission, they are quite uncommon. The genetic diversity of dengue viruses is specific to certain regions, suggesting that the establishment of the virus in new areas is relatively rare, even as dengue has appeared in new regions in recent decades 29.
**Inhibition of the Human Immune System by the Dengue Virus**
The dengue virus disrupts the innate immune response in several ways, as outlined below.
Inhibition of Interferon Signaling by Blocking Signal Transducer: **NS4B**: This protein is linked to the endoplasmic reticulum and may inhibit the phosphorylation of STAT1 following stimulation by type I interferons (alpha and beta). The presence of the dengue virus reduces the activity of Tyk2 kinase, leading to decreased phosphorylation of STAT1. As a result, the innate immune response may be hindered, preventing the production of interferon-stimulated genes (ISGs). Additionally, the cofactors NS2A and NS4A may also contribute to the inhibition of STAT1.
**NS5**: This protein is involved in the inactivation of STAT2, which occurs through the signaling pathway activated by interferons when NS5 is expressed alone. When NS5 is cleaved by the NS2B3 protease, along with NS4B, it can degrade STAT2. Following this cleavage, NS5 associates with E3 ligase, which then targets STAT2 for degradation 30.
Inhibition of the Type I Interferon Response: The NS2B3 protease complex is a proteolytic core made up of the last 40 amino acids of NS2B and the first 180 amino acids of NS3. Cleaving the NS2B3 precursor activates this protease complex.
It plays a role in inhibiting the production of type I interferon by decreasing the activity of the IFN-β promoter. Research has indicated that the NS2B3 protease complex is involved in inhibiting the phosphorylation of IRF3. A recent study has found that the NS2B3 protease complex also cleaves the MITA protein, which is necessary for activating IRF3 31.
Drugs used in the Treatment of Dengue:
Acetaminophen: The exact mechanism of action of acetaminophen, according to its FDA labeling, has not been fully established. However, it is often grouped with NSAIDs (nonsteroidal anti-inflammatory drugs) due to its ability to inhibit the cyclooxygenase (COX) pathways. Acetaminophen is believed to work centrally to reduce pain.
One theory suggests it raises the pain threshold by inhibiting COX-1 and COX-2 isoforms, which are involved in the production of prostaglandins, chemicals that cause pain. Unlike NSAIDs, acetaminophen does not inhibit COX in peripheral tissues, so it lacks anti-inflammatory effects outside the central nervous system. Unlike aspirin, which irreversibly inhibits COX by blocking its active site, acetaminophen appears to inhibit COX indirectly. Research also points to acetaminophen selectively blocking a variant of the COX enzyme, distinct from COX-1 and COX-2, known as COX-3. Its antipyretic (fever-reducing) effects are likely due to its action on the brain's heat-regulating centers, leading to vasodilation, sweating, and heat loss. While the full mechanism is still not fully understood, ongoing research may provide more insights 32.
Metoclopramide: Metoclopramide (MCP) is an antiemetic medication that may also exhibit antiviral effects against dengue virus (DENV) infection. Its mechanism of action against DENV is thought to involve preventing the virus from binding to or entering cells through a pathway mediated by the D2 receptor (D2R). A 2021 study demonstrated that MCP was effective in reducing DENV-induced central nervous system (CNS) neuropathy and mortality in an in-vivo model 33.
Dyphenhydrinate: Dimenhydrinate is a compound consisting of a theoclate salt that breaks down into two active components: diphenhydramine and 8-chlorotheophylline. While the precise mechanism by which dimenhydrinate exerts its effects is not fully established, diphenhydramine, one of its primary components, is thought to play a key role in its therapeutic action. Diphenhydramine is known to alleviate symptoms of nausea, dizziness, and motion sickness, which are often associated with disturbances to equilibrium.
The proposed mechanisms behind diphen-hydramine's effectiveness include its antimuscarinic properties, which involve blocking acetylcholine receptors in the inner ear and the central nervous system, thereby reducing the signals that trigger dizziness and nausea. Additionally, diphen-hydramine functions as an antagonist of histamine H1 receptors, which are involved in the body's response to motion and other stimuli. By blocking these histamine receptors, diphenhydramine helps prevent the symptoms of motion sickness and other related disturbances. Although the exact pathway remains unclear, these actions likely contribute to the drug's ability to restore balance and alleviate nausea 34.
Loratadine: Loratadine exerts its therapeutic effects primarily by targeting H1 histamine receptors. These receptors are present on the surface of a variety of cells, including epithelial cells, endothelial cells, eosinophils, neutrophils, airway cells, and vascular smooth muscle cells, among others. When loratadine binds to these H1 receptors, it blocks the action of histamine, a chemical responsible for triggering allergic responses such as inflammation, swelling, and irritation.
By inhibiting histamine’s interaction with its receptors, loratadine helps to alleviate symptoms of allergies, such as sneezing, itching, and runny nose. Additionally, its effect on airway and vascular smooth muscle cells helps reduce the constriction of airways and the leakage of fluid from blood vessels, further contributing to the relief of allergy symptoms. Because loratadine is a selective H1 receptor antagonist, it is generally less likely to cause sedation compared to first-generation antihistamines 35.
Chloroquine: Chloroquine, a well-known drug, exhibits bactericidal properties against Bacillus megaterium, a type of bacteria. The primary mechanism by which chloroquine exerts its antimicrobial effects involves the inhibition of DNA and RNA biosynthesis within the bacterial cell. This disruption of nucleic acid synthesis leads to the rapid degradation of ribosomes and the breakdown of ribosomal RNA, which are critical components for protein production and cellular function. In addition to its direct impact on nucleic acids, chloroquine also interferes with protein synthesis. This effect, however, appears to be secondary to the drug's primary action of inhibiting DNA replication. The inhibition of DNA replication is believed to be a key step in the bactericidal action of chloroquine, as it prevents the bacteria from replicating its genetic material, thereby halting its ability to grow and divide. The overall mechanism of chloroquine’s antimicrobial action can thus be understood as a multifaceted disruption of essential cellular processes, primarily targeting the synthesis of DNA and RNA, which leads to the subsequent degradation of ribosomes and inhibition of protein synthesis. This combination of effects ultimately results in the death of the bacterial cell 36.
Doxycycline: An antibiotic, inhibits bacterial protein synthesis by interacting with the 30S ribosomal subunit of prokaryotic cells. It does so through an allosteric binding mechanism, meaning that it binds to a site on the ribosome distinct from the active site, leading to a conformational change that interferes with its normal function. This binding prevents the ribosome from properly assembling the components needed for protein synthesis. The specific action of doxycycline involves blocking the interaction between the ribosome and the charged aminoacyl-tRNA (aa-tRNA), which is crucial for protein elongation. The ribosomal A site, part of the mRNA-ribosome complex, serves as the docking site for the aminoacyl-tRNA, where the tRNA brings in the corresponding amino acid to be added to the growing polypeptide chain during translation. By preventing the aminoacyl-tRNA from binding to the A site, doxycycline effectively halts the elongation of the protein chain. As a result, doxycycline disrupts the process of translation, preventing the bacteria from synthesizing the proteins necessary for their growth and reproduction. This inhibition of protein synthesis slows bacterial growth, ultimately leading to the cessation of bacterial proliferation. While doxycycline is primarily bacteriostatic (it inhibits bacterial growth without directly killing the bacteria), it still allows the host immune system to clear the infection by impeding the bacteria's ability to multiply 37.
Zanamivir: Zanamivir is an antiviral medication that functions as a neuraminidase inhibitor, meaning it interferes with the activity of the neuraminidase enzyme, which is crucial for the influenza virus to propagate and spread within the host. Neuraminidase is a protein found on the surface of the influenza virus, and it plays a vital role in the virus’s ability to release new viral particles from infected cells. After a virus infects a host cell and replicates, the neuraminidase enzyme helps the newly formed viral particles detach from the surface of the infected cell, allowing them to spread and infect neighboring cells. By inhibiting the neuraminidase enzyme, zanamivir effectively prevents this process, blocking the virus’s ability to release and spread its new viral particles. This action slows down or stops the spread of the virus throughout the body, helping to limit the severity and duration of the infection. In this way, zanamivir helps to control the influenza virus by interfering with its ability to propagate, giving the immune system a better chance to fight the infection and reducing the risk of further viral transmission 38.
Ivermectin: Ivermectin is an antiparasitic drug that works through a multi-faceted mechanism, making it highly effective against a range of parasitic infections. Its primary action is to target glutamate-gated chloride channels, which are present in the nerve and muscle cells of many parasites. When ivermectin binds to these channels, it increases the flow of chloride ions into the cells, leading to the hyperpolarization of the cell membrane. This disrupts normal neural transmission by inhibiting the ability of the nerve cells to fire properly, which results in paralysis of the parasite. As a consequence of this paralysis, the parasite is unable to maintain vital functions, leading to its eventual death. In addition to its antiparasitic effects, ivermectin has demonstrated anti-inflammatory and antiviral properties 39.
It can inhibit the production of cytokines, which are signaling molecules that play a key role in the inflammatory response. By reducing cytokine production, ivermectin helps to modulate the host’s immune response, leading to less inflammation and tissue damage. This can be particularly beneficial in treating conditions where inflammation contributes to disease progression, such as in certain parasitic or viral infections. Moreover, ivermectin has been shown to inhibit viral replication in some cases.
Its antiviral effects are still being studied, but it has been found to interfere with the ability of certain viruses to replicate within host cells, further expanding its therapeutic potential. Ivermectin's action involves a variety of cellular pathways, making it a versatile drug with applications beyond parasitology. It is effective against a broad range of parasites, including roundworms, lice, mites, and other ectoparasites. Additionally, its antiviral and immunomodulatory effects suggest that ivermectin may have potential applications in treating viral infections and conditions characterized by excessive inflammation. Its unique combination of mechanisms makes ivermectin an important drug not only for parasitic infections but also for potential use in other therapeutic contexts 40.
Oral Rehydration Salt: Oral rehydration salts (ORS) are a simple yet highly effective treatment for replenishing lost fluids and electrolytes, particularly in cases of dehydration caused by vomiting or diarrhea. The main components of ORS glucose, sodium, potassium, and water work together to rapidly restore the body’s fluid balance and prevent dehydration, a condition that can be life-threatening, especially in vulnerable populations such as infants and young children 41. The mechanism of action of ORS revolves around its ability to enhance fluid absorption in the intestines. Here's how each key ingredient contributes:
Glucose: The addition of glucose in ORS plays a crucial role in the absorption of both sodium and water in the small intestine. Glucose stimulates the sodium-glucose co-transporter, a mechanism that actively transports sodium and glucose into the intestinal cells. This process increases the osmotic gradient, allowing more water to be absorbed along with sodium. This helps the body retain fluids more effectively, rehydrating the body more quickly.
Potassium Chloride: Potassium is an essential electrolyte that helps maintain the body's fluid and electrolyte balance. Diarrhea often leads to a significant loss of potassium, which can result in hypokalemia (low potassium levels). Potassium chloride in ORS helps prevent this condition by replenishing potassium levels, supporting proper muscle function, and helping regulate the heart's rhythm.
Sodium Citrate: Sodium citrate acts to prevent acidosis, a common complication of diarrhea, where the blood becomes too acidic. During diarrhea, the body loses not only water but also bicarbonate (a base), leading to an acidic imbalance. Sodium citrate, by providing a source of bicarbonate, helps to neutralize the excess acid in the body and maintain a proper pH balance, thereby preventing or correcting acidosis 42.
The combination of these ingredients in ORS allows the intestines to absorb fluids more efficiently, replacing the water and electrolytes lost due to diarrhea or vomiting. This is particularly important in the treatment of dehydration, which is a frequent and dangerous consequence of severe diarrhea, especially in young children.
ORS is often used to treat and prevent dehydration from gastroenteritis or other illnesses that cause diarrhea, and it is widely recommended by health organizations such as the World Health Organization (WHO) for its effectiveness and simplicity. In summary, ORS works by promoting the rapid absorption of fluids and electrolytes, correcting imbalances that can occur due to dehydration, and preventing complications like hypokalemia and acidosis. Its use is a cornerstone in the management of dehydration caused by diarrhea, offering a life-saving treatment that is easy to administer, affordable, and accessible, especially in resource-limited settings 43.
Food Supplements for the Treatment:
Coconut Water: Dengue fever often leads to significant dehydration, which can contribute to a range of complications during the illness. To help manage this, it is highly beneficial to incorporate coconut water into your daily diet. Coconut water is naturally rich in electrolytes, particularly potassium and sodium, which are essential for replenishing lost fluids and maintaining hydration. This helps to restore the body's fluid balance, supporting overall health and reducing the risk of further complications.
In addition to its hydrating properties, coconut water also aids in detoxifying the body. It helps flush out toxins, promoting kidney function and ensuring that harmful substances are removed efficiently. By keeping your body hydrated and supporting its natural detoxification processes, coconut water can help you feel more energized and healthier for longer periods, especially during recovery from dengue. Thus, making coconut water a part of your daily routine can be a simple yet effective way to support your body's healing process 44.
Kiwi: Kiwi is often highly recommended as a beneficial fruit to consume during dengue recovery due to its numerous health benefits. This small yet nutrient-dense fruit is packed with essential vitamins and minerals, making it an excellent addition to the diet during illness. Kiwi is particularly rich in Vitamin E and Vitamin A, both of which play crucial roles in boosting the immune system and promoting healing. Vitamin E is known for its antioxidant properties, which help protect the body from oxidative stress, while Vitamin A supports cellular repair and strengthens immune function. In addition to vitamins, kiwi is a good source of potassium, an important mineral that helps regulate fluid balance in the body and supports proper muscle and nerve function. During dengue, the body is at risk of dehydration due to fluid loss, and potassium helps to replenish the body’s electrolyte levels, preventing further complications like muscle cramps or weakness. Kiwi also plays a role in cardiovascular health by helping to manage high blood pressure and hypertension. The fruit's natural compounds help relax blood vessels, which can contribute to more stable blood pressure levels. This is particularly beneficial for individuals recovering from dengue, as maintaining healthy circulation and heart function is vital for overall recovery. Most importantly, kiwi supports the production of collagen, a vital protein for tissue repair and wound healing. Collagen is essential for the formation of new blood vessels and the recovery of damaged tissues, which is particularly important for dengue patients who may experience a decrease in platelet count and blood vessel integrity 45.
Overall, including kiwi in the diet during dengue recovery can aid in immune support, help regulate electrolyte balance, manage blood pressure, and promote tissue repair, all of which contribute to a smoother and faster recovery process.
Broccoli: Broccoli, often disliked during childhood, is a powerhouse vegetable that offers numerous health benefits, particularly when it comes to recovery from illnesses like dengue. One of its standout qualities is its high content of Vitamin K, a nutrient that plays a crucial role in blood clotting and platelet regeneration. During dengue, where a drop in platelet count is a common concern, including broccoli in the diet can help support the body’s ability to regenerate platelets, thereby aiding in the recovery process.
Beyond Vitamin K, broccoli is rich in a variety of essential minerals, including calcium, magnesium, and iron, which contribute to overall health and support bodily functions. The vegetable is also packed with antioxidants like Vitamin C, which helps strengthen the immune system, reduce inflammation, and protect cells from damage. These antioxidants are particularly important during illness, as they help the body recover more quickly and reduce oxidative stress caused by infections. Additionally, broccoli’s high fiber content supports digestive health, while its low-calorie, nutrient-dense profile makes it an ideal choice for maintaining a healthy diet during recovery. Whether added to salads, pasta dishes, or enjoyed as a side, broccoli is a versatile and highly beneficial vegetable that should definitely be included in a dengue recovery diet for its ability to support immune function, blood platelet regeneration, and overall healing 46.
Spinach: Green vegetables have long been celebrated for their exceptional nutritional value, and spinach stands out as one of the healthiest choices among them. Packed with a wide array of vitamins, minerals, and essential nutrients, spinach is a powerhouse for boosting overall health.
One of its key benefits is its high content of iron, an essential mineral that plays a vital role in the production of red blood cells. Adequate iron intake is particularly important during illness, as it supports the body’s ability to regenerate blood cells and maintain healthy circulation. For those recovering from dengue, where low platelet and red blood cell counts are a concern, spinach can help promote the production of healthy blood cells and boost overall vitality.
In addition to iron, spinach is also a rich source of omega-3 fatty acids, which are known for their anti-inflammatory properties and their ability to support the immune system. Omega-3s help regulate inflammation in the body, making them especially beneficial during recovery from infections like dengue, where the immune system may be under stress. These healthy fats also support brain function, cardiovascular health, and help maintain the body’s overall energy levels.
Furthermore, spinach is a great source of Vitamin C, Vitamin A, and folate, all of which play important roles in strengthening the immune system, enhancing the body’s natural defenses, and aiding in the repair of damaged tissues. The antioxidants present in spinach also help fight oxidative stress and protect cells from damage caused by illness. Given its high nutrient density and ability to promote blood health and immunity, spinach should be a regular part of the diet, especially during dengue recovery. Whether consumed raw in salads, sautéed, or blended into smoothies, spinach is a versatile and powerful vegetable that supports healing, boosts platelet count, and strengthens the body's defenses 47.
Berries: Berries such as raspberries, goji berries, blackberries, blueberries, and strawberries are not only delicious but also powerhouses of nutrition, particularly when it comes to boosting platelet count. These fruits are rich in antioxidants, which are compounds that help protect the body from oxidative stress and cellular damage. The high concentration of antioxidants in berries, including polyphenols, plays a key role in supporting overall health and recovery, especially in conditions like dengue, where platelet count can be dangerously low.
Clinical studies have shown that the moderate consumption of berries can have a positive impact on platelet count, making them an excellent addition to the diet for those seeking to support their recovery. Berries are known to promote the production of platelets, the blood cells responsible for clotting, which is particularly important during dengue, as the virus can cause a significant drop in platelet levels. The polyphenols found in berries are believed to stimulate the production of these blood cells, thereby helping to restore balance and promote healing.
In addition to their benefits for platelet production, berries are also known for their positive effects on HDL cholesterol (often referred to as "good cholesterol"). Regular consumption of berries has been shown to help increase HDL levels, which play a crucial role in reducing the risk of cardiovascular disease by helping to clear excess cholesterol from the bloodstream.
Moreover, berries are packed with vitamins, particularly Vitamin C, which supports immune function and aids in the absorption of iron, an essential nutrient for the production of healthy blood cells. The fiber in berries also supports digestive health, helping to maintain overall well-being during recovery.
Overall, incorporating a variety of berries into the diet can provide numerous health benefits, from improving platelet count to supporting cardiovascular health and boosting immunity. Whether eaten fresh, added to smoothies, or sprinkled over salads and cereals, berries are a tasty and nutritious way to support the body’s healing process, especially during illness 48.
Carrots and Beetroots: These vegetables are rich in a wide array of antioxidants, vitamins, and minerals, making them some of the best foods to support overall health and particularly to help increase platelet counts. When recovering from conditions like dengue, where platelet levels may drop significantly, these nutrient-dense vegetables can play a key role in the recovery process. One of the standout nutrients found in many of these vegetables is folate (also known as Vitamin B9). Folate is essential for the production of healthy blood cells, including red blood cells and platelets. It helps in the formation and maturation of new blood cells, thereby contributing to the restoration of a balanced blood count. This makes it particularly important for those recovering from illnesses that affect blood cell production, such as dengue, where low platelet count is a major concern. In addition to folate, these vegetables are packed with a variety of antioxidants, such as vitamin C, beta-carotene, and flavonoids, which help protect cells from oxidative damage caused by free radicals. This antioxidant activity is crucial during recovery, as it aids in reducing inflammation and supports the immune system by neutralizing harmful molecules that can damage tissues and slow down the healing process.
The combination of folate and antioxidants in these vegetables helps not only to enhance platelet production but also to promote overall cellular health, improve immune function, and reduce the risk of further complications. Vegetables such as spinach, kale, broccoli, and others are especially beneficial for replenishing nutrients and strengthening the body's natural defenses, which is why they are often recommended as part of a recovery diet. By including a variety of these vegetables in your daily meals, you provide your body with the essential nutrients it needs to heal more effectively, maintain healthy blood cell production, and protect against cellular damage, ultimately contributing to a faster and smoother recovery 49.
Pumpkin: Pumpkin is a highly nutritious food that can be particularly beneficial for increasing platelet count, thanks to its rich content of Vitamin A. This essential nutrient plays a critical role in supporting the production of platelets in the bone marrow, the site where blood cells are produced. Vitamin A helps to regulate the growth and development of blood cells, including platelets, making it an important dietary component for individuals with low platelet levels, such as those recovering from illnesses like dengue. By boosting platelet production, pumpkin can help maintain a healthy blood cell balance and support overall recovery. In addition to Vitamin A, pumpkin is packed with other essential nutrients, including vitamin C, fiber, and potassium, all of which contribute to immune health, digestive function, and overall well-being. These nutrients work together to support the body’s natural healing processes, making pumpkin an excellent addition to a balanced diet for those looking to improve their platelet count. However, if you are pregnant and seeking ways to increase your platelet count, it’s important to approach dietary changes with caution. While pumpkin and other nourishing foods can support overall health and platelet production, it’s crucial to consult with your doctor or healthcare provider before making any significant changes to your diet. Pregnancy requires special consideration when it comes to nutrition, and your doctor can provide personalized advice to ensure that you are supporting both your health and the health of your baby in the best possible way. By working with your healthcare provider, you can make informed choices about how to incorporate foods like pumpkin into your diet, helping to boost platelet production while also ensuring a healthy, balanced pregnancy 50.
Nutrients that Increases Platelet Count: Several key nutrients play an important role in increasing platelet count, supporting overall blood health, and aiding recovery from conditions like dengue or other illnesses that cause low platelet levels. Here's a breakdown of the most beneficial nutrients:
Vitamin C: This vitamin is essential for the health of blood vessels and the immune system. It helps in the absorption of iron, an important mineral for blood cell production, and aids in the overall formation of platelets. Foods rich in Vitamin C, such as citrus fruits, strawberries, bell peppers, and leafy greens, can support platelet production by enhancing the body’s ability to produce healthy blood cells.
Vitamin K: Vitamin K is crucial for blood clotting, and it plays a role in regulating platelet activity.
It helps with the formation of proteins involved in blood clotting and is vital for maintaining a stable platelet count. Green leafy vegetables like spinach, kale, and broccoli are rich in Vitamin K and can aid in platelet production.
Folate (Vitamin B9): Folate is a B-vitamin that is essential for the production of red blood cells and platelets. It supports the bone marrow in producing new blood cells, which is critical when platelet levels are low. Foods such as lentils, spinach, asparagus, and avocados are excellent sources of folate.
Iron: Iron is a vital mineral for the production of hemoglobin and the overall function of red blood cells. Iron helps maintain a healthy level of red blood cells, indirectly supporting platelet production. Foods like red meat, beans, fortified cereals, and leafy greens are high in iron and can contribute to raising platelet levels.
Omega-3 Fatty Acids: Found in foods like fish (salmon, mackerel), flaxseeds, and walnuts, omega-3 fatty acids help reduce inflammation and support the immune system. These healthy fats can enhance platelet function and overall cardiovascular health, promoting better circulation and platelet activity.
Vitamin B12: Vitamin B12 is essential for the healthy development of blood cells, including platelets. A deficiency in B12 can lead to anemia and a low platelet count. Foods such as eggs, dairy, fortified cereals, and meat are good sources of Vitamin B12.
Zinc: Zinc plays a significant role in immune function and wound healing. It also helps in the production of new blood cells, including platelets. Zinc-rich foods include oysters, meat, beans, nuts, and seeds.
Antioxidants: Nutrients like Vitamin E and beta-carotene (found in carrots, sweet potatoes, and other colorful vegetables) are antioxidants that help protect the body from oxidative stress. They support the immune system and protect blood cells, including platelets, from damage 51-52. A balanced diet rich in these nutrients can help promote the production of platelets and support overall blood health. While these nutrients are important for platelet production, it’s essential to consult with a healthcare provider for personalized guidance, especially if you are dealing with a specific medical condition.
Emerging Therapy for Dengue:
Monoclonal Antibodies:
PRV-002 Monoclonal Antibody: PRV-002 is a monoclonal antibody aimed at neutralizing the dengue virus by targeting its envelope protein. Early preclinical studies have indicated that it can reduce viral replication and lessen the severity of infection in animal models. Ongoing clinical trials are assessing its safety and efficacy in humans.
DEN-80 Monoclonal Antibody: DEN-80 is another monoclonal antibody designed to target the dengue virus. It binds to a conserved area of the virus's envelope protein, blocking viral replication. Laboratory studies suggest it could be effective against all four dengue serotypes, offering promise in reducing disease severity.
Combination Antibody Therapy: Researchers are investigating the use of multiple monoclonal antibodies targeting different regions of the dengue virus. Preliminary research indicates that combining these antibodies may provide broader protection, potentially improving outcomes for patients infected with different strains of the virus.
Monoclonal Antibodies for Severe Dengue: For severe dengue cases, where inflammation and blood vessel leakage are critical concerns, certain monoclonal antibodies are being tested to help modulate the immune response. These antibodies have shown potential in reducing harmful inflammation, which may help prevent complications such as hemorrhagic fever.
Humanized Monoclonal Antibodies: To enhance safety and effectiveness, scientists are developing humanized monoclonal antibodies, which are less likely to provoke immune rejection. One such antibody targeting the dengue E protein has shown promising results in early trials.
Immunomodulatory Effects: Some monoclonal antibodies are designed not only to neutralize the virus but also to regulate the immune response, which is crucial in severe dengue. This approach could help reduce viral load and prevent excessive immune activation, which could otherwise lead to complications like shock.
Broad-Spectrum Monoclonal Antibodies: Since, dengue exists in four serotypes, immunity to one type does not protect against the others. Researchers are focusing on monoclonal antibodies that target conserved regions of the virus, which could offer protection against all four serotypes.
Post-Exposure Prophylaxis: Monoclonal antibodies could be used as a post-exposure treatment for individuals who have been recently exposed to the virus. Administering these antibodies shortly after exposure may help prevent infection or mitigate the severity of the disease, making it a useful option for high-risk individuals.
Clinical Trials of Monoclonal Antibodies: Clinical trials are essential for assessing the safety and efficacy of monoclonal antibodies in humans. Early-phase trials with antibodies like PRV-002 have shown them to be generally safe, with only mild side effects. More extensive trials are required to fully evaluate their therapeutic potential.
Bispecific Antibodies: Bispecific antibodies, which can target both the dengue virus and the immune cells involved in the disease, are under investigation. These antibodies may provide a more comprehensive treatment approach by neutralizing the virus and simultaneously regulating the immune response that contributes to severe dengue 53-55.
Advancements in Dengue Vaccine Development:
Current Vaccines (Dengvaxia and Qdenga): Dengvaxia and Qdenga are approved vaccines in certain regions, providing protection against dengue. However, Dengvaxia has raised safety concerns, especially for individuals who have not been previously infected with the virus.
Ongoing research is focused on creating universal or tetravalent vaccines that can protect against all four dengue serotypes while being safer for those with no prior exposure to the virus.
Universal and Tetravalent Vaccines: Researchers are working on vaccines that offer protection against all four dengue serotypes. These new vaccines aim to improve immunogenicity (the ability to trigger a strong immune response) and safety, addressing issues seen with earlier vaccines like Dengvaxia.
New Vaccine Candidates (C1, C2, C3): Innovative vaccine candidates, such as C1 and C3, are being explored. These vaccines target specific components of the dengue virus, intending to enhance immune response while minimizing risks. Research is focused on improving their effectiveness and safety profiles.
Ensuring Safety and Efficacy: A key challenge in developing dengue vaccines is ensuring they are both safe and effective. Vaccines must be designed to stimulate a protective immune response without causing enhanced disease (worsening of symptoms in subsequent infections). Researchers are focused on achieving a balanced immune response to prevent severe outcomes.
Long-Term Protection: New vaccines are being developed to offer long-lasting immunity against dengue. There is also ongoing research into the need for booster doses to maintain immunity over time, especially for those who may require additional protection.
Emerging Vaccine Technologies: New vaccine technologies, such as viral vector and mRNA vaccines, are being explored. These platforms provide flexible and rapid methods to develop vaccines that can be adapted to address emerging dengue strains.
Access and Distribution Challenges: A critical aspect of dengue vaccine development is ensuring global access. The vaccines must be affordable, scalable, and easy to distribute, especially in dengue-endemic regions. Efforts are underway to incorporate these vaccines into national immunization programs for widespread use.
Future Directions: Looking ahead, there is optimism that next-generation vaccines will offer improved safety, wider protection, and better distribution capabilities. These vaccines are expected to play a major role in controlling the global spread of dengue 56-58.
Host-Directed Therapies for Dengue: Research into host-targeted treatments for dengue is focused on modulating the body's immune response to prevent severe disease, rather than directly attacking the virus itself. These approaches aim to control inflammation and enhance the function of immune cells.
Controlling Inflammation: Overactive infla-mmation is a major factor in the development of severe dengue, including conditions like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Several strategies are being explored to manage this excessive immune response:
Cytokine Blockade: Monoclonal antibodies and small molecules that target key inflammatory cytokines (such as TNF-α and IL-1β) are being studied. These treatments aim to reduce the damaging effects of inflammation while allowing the immune system to fight the virus effectively.
Complement System Regulation: Inhibiting the complement system, which contributes to tissue damage in severe dengue, may help minimize vascular leakage and organ injury.
NF-κB Pathway Inhibition: Targeting the NF-κB pathway, a critical regulator of inflammation, could help lower excessive cytokine production, potentially reducing severe disease progression 59.
Enhancing Immune Cell Function: A well-balanced immune response is crucial for effectively controlling dengue infection and avoiding severe outcomes. Research is focused on improving the performance of key immune cells:
Macrophage Modulation: Macrophages are essential in both clearing the virus and contributing to inflammation. Therapies that promote an anti-inflammatory macrophage response could help reduce harmful inflammation and prevent the progression to severe disease.
T Cell Regulation: CD8+ T cells are important for viral clearance but can cause tissue damage if overly activated. Researchers are exploring ways to boost regulatory T cells (Tregs), which can suppress harmful T-cell responses and maintain balance in the immune reaction.
TLR Agonists: Activating specific Toll-like receptors, particularly TLR3, may enhance the body's ability to fight the virus while preventing excessive inflammation, ensuring a more controlled immune response 60.
Gene Editing and CRISPR-Cas9 for Dengue Control: Gene-editing technologies, particularly CRISPR-Cas9 and gene-drive systems, offer innovative strategies to control the spread of dengue by either targeting the virus directly or modifying mosquito populations.
CRISPR-Cas9 for Mosquito Gene Editing: CRISPR-Cas9 can be used to modify the genetic makeup of Aedes aegypti mosquitoes, the primary vectors of dengue. Researchers are investigating ways to create mosquitoes that are resistant to the dengue virus or unable to transmit it. By editing genes related to viral replication or immune responses, CRISPR could potentially prevent the spread of the disease.
Gene-Drive Technology: Gene-drive systems aim to rapidly spread genetic modifications throughout wild mosquito populations. This could involve engineering mosquitoes that either cannot carry the dengue virus or produce offspring that are immune to it. Gene-drive technologies hold great potential for reducing dengue transmission by altering mosquito populations on a large scale.
Virus Resistance and Inhibition: Some studies focus on using CRISPR to enhance the immune systems of mosquitoes, making them less vulnerable to the dengue virus or preventing viral replication. These gene-edited mosquitoes could block virus transmission by disrupting the virus's lifecycle within the mosquito.
Ethical and Ecological Considerations: While these technologies show promise, they remain in the experimental phase. There are several challenges, including ethical concerns, ecological impacts, and regulatory issues. Concerns include the potential for unintended consequences, such as disrupting ecosystems or developing resistance to the genetic modifications 61-62.
Advances in Platelet Transfusion and Supportive Care for Severe Dengue: Recent research has made significant strides in improving the supportive care of severe dengue, particularly in the areas of platelet transfusion and fluid management. While these treatments are not cures, they are crucial in preventing complications such as shock and bleeding, which are the leading causes of death in severe cases.
Platelet Transfusion: Early platelet transfusion is vital in preventing bleeding, particularly when platelet counts fall below critical thresholds. Studies suggest that transfusions should be based on clinical symptoms rather than just platelet counts, as this can help reduce the risk of hemorrhage and improve patient outcomes.
Personalized Fluid Resuscitation: New approaches to fluid therapy focus on individualized treatment plans, adjusting fluid intake based on the patient’s specific needs, such as blood pressure, heart rate, and urine output. This personalized approach helps avoid complications like fluid overload and dehydration, ultimately improving survival rates.
Better Patient Outcomes: Combining early platelet transfusions with personalized fluid management has shown promising results in reducing complications and enhancing recovery in severe dengue patients, highlighting the effectiveness of tailored interventions.
Challenges and Considerations: Despite these advances, careful monitoring is essential to avoid overuse or inappropriate application of treatments. Unnecessary transfusions or improper fluid administration can lead to adverse effects, underscoring the need for vigilant management 63-64.
DISCUSSION: The ongoing global health threat posed by dengue virus (DENV) underscores the urgent need for enhanced understanding and effective strategies to combat this disease. This report aims to synthesize current research, identify critical gaps in knowledge, and guide future efforts to address the public health challenges posed by DENV. Through a comprehensive exploration of various aspects of dengue virus biology, transmission, pathogenesis, and control measures, this paper highlights the complexity of the disease and the pressing need for integrated, multidisciplinary approaches to mitigate its impact. One of the key insights from the literature reviewed is the intricate evolutionary history and epidemiology of DENV. The virus’s transition from a sylvatic cycle to widespread urban transmission, facilitated by its mosquito vectors, has been a significant factor in its global spread. Understanding the evolutionary dynamics of DENV, particularly the interactions between the virus and its mosquito vectors, is crucial for developing strategies to prevent transmission. The adaptability of DENV to changing environmental conditions and its ability to evolve rapidly complicates efforts to control outbreaks. Therefore, a better understanding of these evolutionary processes could inform more targeted interventions, such as the development of mosquito control strategies or genetically modified mosquito populations.
Furthermore, the investigation into viral replication and the role of untranslated regions (UTRs) in regulating genome synthesis and translation has revealed significant insights into the molecular mechanisms underpinning DENV biology. These regions play a crucial role in the virus's ability to replicate efficiently, and their manipulation could potentially lead to new antiviral strategies. However, as the virus evolves, so too must our understanding of these mechanisms. This ongoing need for research underscores the complexity of DENV and highlights the challenges in identifying targets for therapeutic intervention that can remain effective against evolving strains of the virus.
The pathogenesis of dengue illness and the host immune response to DENV infection are equally critical areas of research. The severity of dengue disease, ranging from mild symptoms to potentially fatal hemorrhagic fever or dengue shock syndrome, is influenced by various host and virus determinants. The immune system’s response to infection, particularly the phenomenon of antibody-dependent enhancement (ADE), plays a central role in determining the outcome of the disease. Insights into host-pathogen interactions, including the identification of key host immune factors that influence susceptibility and disease severity, could lead to novel therapeutic and preventive approaches. The development of animal models to study these interactions is essential, as it allows for a better understanding of the disease's progression and the identification of potential therapeutic targets. One of the challenges in controlling dengue is the complexity of the virus’s interaction with its mosquito vectors. DENV-mosquito interactions are multifaceted, with environmental and genetic factors influencing the ability of mosquitoes to transmit the virus. The spatial and temporal dynamics of dengue transmission also add layers of complexity, making it difficult to predict and prevent outbreaks. A more detailed understanding of these interactions, including the role of mosquito populations in viral spread and the environmental factors that affect transmission, could significantly enhance vector control strategies. Furthermore, the emergence of new DENV serotypes, as well as the ongoing adaptation of the virus to new ecological niches, poses additional challenges in managing the spread of the disease. The epidemiology of dengue, including the spatial and temporal dynamics of transmission, remains an area of considerable concern. Despite ongoing efforts to improve surveillance and early warning systems, the unpredictability of dengue outbreaks in some regions is a major hurdle.
Advances in data analytics, satellite monitoring, and machine learning could help improve predictive models and enable more proactive responses to potential outbreaks. Additionally, the resurgence of dengue in areas that had previously seen a decline emphasizes the need for sustained vigilance and ongoing research into factors that influence the resurgence of transmission, such as urbanization, climate change, and population mobility. In response to the growing burden of dengue, significant progress has been made in the development of vaccines, antiviral drugs, and passive immunotherapy. The success of the Dengvaxia vaccine, despite its limitations, has highlighted the potential for vaccination as a tool in reducing disease burden. However, concerns over safety, efficacy, and long-term effectiveness require further investigation before widespread implementation can occur. Meanwhile, novel antiviral compounds and therapies are under investigation, offering the possibility of more targeted treatments for dengue. The ongoing development of monoclonal antibodies and other immunotherapies also shows promise, particularly in providing a short-term solution for individuals at high risk of severe disease.
CONCLUSION: Dengue fever has emerged as a significant global health threat, increasingly affecting many nations around the world. This mosquito-borne viral infection, characterized by sudden high fever, severe headaches, pain behind the eyes, joint pain, rash, and in some cases, bleeding, has become a growing concern for public health officials. The disease is transmitted primarily by the Aedes aegypti mosquito, and the rising prevalence of dengue can be attributed to various factors, including urbanization, climate change, and increased global travel. Consequently, governments and pharmaceutical industries have been intensifying their efforts to develop innovative strategies for improving both the diagnosis and treatment of dengue.
However, the real challenge lies not only in the creation of these new techniques but also in their effective implementation. The ability to rapidly and efficiently introduce and adopt these strategies into healthcare systems, especially in regions with limited resources, is crucial to controlling the spread of dengue. Although there have been significant advancements in medical research and drug development, the effectiveness of these innovations will depend on how well they are integrated into routine healthcare practices. Additionally, countries with high dengue burdens often face challenges related to insufficient healthcare infrastructure, inadequate training for medical personnel, and a lack of awareness among the general population.
In order to make meaningful progress in the fight against dengue, there is an urgent need to increase global awareness and educate the public about preventive measures. Prevention is at the core of dengue control, as there is currently no specific antiviral treatment for the disease. Preventive strategies primarily focus on controlling the mosquito population and avoiding mosquito bites. These can be achieved through a combination of measures, such as the use of insect repellent, the installation of mosquito nets, and the elimination of mosquito breeding sites. Public health campaigns must emphasize the importance of these measures and encourage individuals to take personal responsibility for their role in curbing the spread of the disease. Furthermore, international collaboration is key to addressing the dengue problem on a global scale. Since the disease is prevalent in many parts of the world, particularly in tropical and subtropical regions, there is a need for cooperative efforts between governments, the healthcare sector, international organizations, and the private sector. Governments should priorities funding for dengue research, improve surveillance systems to monitor outbreaks, and strengthen vector control programs. International organizations, such as the World Health Organization (WHO), play a critical role in coordinating efforts, providing technical expertise, and facilitating the sharing of knowledge and resources across borders. The pharmaceutical industry must continue to invest in the development of vaccines and treatments, as well as in the production of diagnostic tools that can help detect the virus at an early stage.
The role of local communities in the fight against dengue cannot be overstated. Individuals must be encouraged to adopt preventive practices, such as clearing standing water from containers where mosquitoes breed, using mosquito repellent, and wearing protective clothing. Community-based initiatives can also be effective in raising awareness and engaging people in mosquito control efforts. For instance, local governments can collaborate with community leaders to organize neighborhood clean-up campaigns to eliminate mosquito breeding grounds. Such grassroots efforts, when combined with government and global initiatives, can have a significant impact on reducing the incidence of dengue.
In addition to these preventive measures, advancements in medical research and healthcare technologies are helping to improve the diagnosis and treatment of dengue. A major breakthrough in recent years has been the development of a dengue vaccine, which, although not universally available, has shown promising results in reducing the severity of the disease. Moreover, improved diagnostic tools, such as rapid diagnostic tests, allow for faster identification of dengue cases, enabling timely treatment and reducing the risk of complications. The development of antiviral drugs remains an area of active research, with the goal of providing a specific treatment for dengue. However, given that the disease is caused by multiple serotypes of the dengue virus, finding a universally effective treatment remains a significant challenge.
In conclusion, tackling the global threat of dengue requires a multi-faceted approach that combines effective medical interventions, public health initiatives, and individual responsibility. Governments, healthcare organizations, and pharmaceutical companies must work together to ensure the successful implementation of strategies aimed at improving diagnosis, treatment, and prevention. At the same time, raising awareness and educating the public about the importance of dengue prevention is essential in controlling the disease's spread. By fostering collaboration at all levels, from local communities to international bodies, it is possible to reduce the burden of dengue and protect vulnerable populations worldwide. Ultimately, only through combined efforts and sustained commitment can we hope to curb the rising prevalence of this dangerous disease.
ACKNOWLEDGEMENT: Nil
CONFLICT OF INTEREST: Nil
REFERENCES:
- Guzman MG and Harris E: Dengue. The Lancet 2015; 385(9966): 453-65. doi.org/ 10.1016/S0140-6736(14)60572-9
- Balmaseda A, Standish K, Mercado JC, Matute JC, Tellez Y, Saborío S, Hammond SN, Nuñez A, Avilés W, Henn MR and Holmes EC: Trends in patterns of dengue transmission over 4 years in a pediatric cohort study in Nicaragua. The Journal of Infectious Diseases 2010; 201(1): 5-14.doi.org/10.1086/648592
- Endy TP, Anderson KB, Nisalak A, Yoon IK, Green S, Rothman AL, Thomas SJ, Jarman RG, Libraty DH and Gibbons RV: Determinants of inapparent and symptomatic dengue infection in a prospective study of primary school children in Kamphaeng Phet, Thailand. PLoS Neglected Tropical Diseases 2011; 5(3): 975.doi.org/10.1371/journal.pntd.0000975
- Gupta N, Srivastava S, Jain A and Chaturvedi UC: Dengue in India. Indian Journal of Medical Research 2012; 136(3): 373-90.
- Bandyopadhyay S, Lum LC and Kroeger A: Classifying dengue: a review of the difficulties in using the WHO case classification for dengue haemorrhagic fever. Tropical Medicine & International Health 2006; 11(8): 1238-55. doi.org/10.1111/j.1365-3156.2006.01678.x
- Mousson L, Dauga C, Garrigues T, Schaffner F, Vazeille M and Failloux AB: Phylogeography of Aedes (Stegomyia) aegypti (L.) and Aedes (Stegomyia) albopictus (Skuse) (Diptera: Culicidae) based on mitochondrial DNA variations. Genetics Research 2005; 86(1): 1-1 doi.org/10.1017/S0016672305007627
- Lambrechts L, Scott TW and Gubler DJ: Consequences of the expanding global distribution of Aedes albopictus for dengue virus transmission. PLoS neglected tropical diseases 2010; 4(5): e646.doi.org/10.1371/journal.pntd.0000646
- WHO Dengue: Guidelines for diagnosis, treatment, prevention, and control in sub-Saharan Africa and 13 countries in South America. 2009 Geneva World Health Organization.
- World Health Organization, Special Programme for Research, Training in Tropical Diseases, World Health Organization. Department of Control of Neglected Tropical Diseases, World Health Organization. Epidemic, Pandemic Alert. Dengue: guidelines for diagnosis, treatment, prevention and control. World Health Organization; 2009
- World Health Organization. Comprehensive guideline for prevention and control of dengue and dengue haemorrhagic fever.
- Cleland JB and Bradley B: Further experiments in the etiology of dengue fever. Epidemiology & Infection 1919; 18(3): 217-54. doi.org/10.1017/S0022172400007476
- McLaughlin JW: Researches into the etiology of dengue. Journal of the American Medical Association 1886; 6(25): 673-80. doi:10.1001/jama.1886.04250060057001
- Carrington LB and Simmons CP: Human to mosquito transmission of dengue viruses. Frontiers in Immunology 2014; 5: 29. doi.org/10.3389/fimmu.2014.00290
- Mulik V, Dad N and Buhmaid S: Dengue in pregnancy. European Journal of Obstetrics & Gynecology and Reproductive Biology 2021; 261: 205 doi.org/10.1016/j.ejogrb.2021.04.035
- Harris WH and Duval CW: Studies upon the Etiology of Dengue Fever: I. Experimental Transmission to the Lower Animal. The Journal of Experimental Medicine 1924; 40(6): 817-33.doi.org/10.1084/jem.40.6.817
- Murray NE, Quam MB and Wilder-Smith A: Epidemiology of dengue: past, present and future prospects. Clinical Epidemiology 2013; 299-309. doi.org/10.2147/CLEP.S34440
- Chen WJ: Dengue outbreaks and the geographic distribution of dengue vectors in Taiwan: a 20-year epidemiological analysis. Biomedical Journal 2018; 41(5): 283- doi.org/10.1016/j.bj.2018.06.002
- Thai KT and Anders KL: The role of climate variability and change in the transmission dynamics and geographic distribution of dengue. Experimental Biology and Medicine 2011; 236(8): 944-54. doi.org/10.1258/ebm.2011.010402
- Jing Q and Wang M: Dengue epidemiology. Global Health Journal 2019; 3(2): 37-45. doi.org/10.1016/j.glohj.2019.06.002
- Thomas SJ, Strickman D and Vaughn DW: Dengue epidemiology: virus epidemiology, ecology, and emergence. Advances in Virus Research 2003; 61: 235-89. doi.org/10.1016/S0065-3527(03)61006-7
- Grange L, Simon-Loriere E, Sakuntabhai A, Gresh L, Paul R and Harris E: Epidemiological risk factors associated with high global frequency of inapparent dengue virus infections. Frontiers in Immunology 2014; 5: 280. doi.org/10.3389/fimmu.2014.00280
- Andraud M, Hens N, Marais C and Beutels P: Dynamic epidemiological models for dengue transmission: a systematic review of structural approaches. PloS one 2012; 7(11): 49085 doi.org/10.1371/journal.pone.0049085
- Roy SK and Bhattacharjee S: Dengue virus: epidemiology, biology, and disease aetiology. Canadian Journal of Microbiology 2021; 67(10): 687-702.
- Andraud M, Hens N, Marais C and Beutels P: Dynamic epidemiological models for dengue transmission: a systematic review of structural approaches. PloS one 2012; 7(11): 49085.doi.org/10.1371/journal.pone.0049085
- Wearing HJ and Rohani P: Ecological and immunological determinants of dengue epidemics. Proceedings of the National Academy of Sciences 2006; 103(31): 11802-7. doi.org/10.1073/pnas.0602960103
- Sandesh B, Mohiniraj A, Bharat C and Balasheb G: Issue 2 www.jetir.org (ISSN-2349-5162). JETIR2302576 Journal of Emerging Technologies and Innovative Research [Internet]. 2023 [cited 2024 Sep 18]; 10. Available from: https://www.jetir.org/papers/JETIR2302576.pdf
- Xi Z, Ramirez JL and Dimopoulos G: The Aedes aegypti toll pathway controls dengue virus infection. PLoS Pathogens 2008; 4(7): 1000098. doi.org/10.1371/journal.ppat.1000098
- Martina BE, Koraka P and Osterhaus AD: Dengue virus pathogenesis: an integrated view. Clinical Microbiology Reviews 2009; 22(4): 564-81.doi.org/10.1128/cmr.00035-09
- Chuansumrit A and Tangnararatchakit K: Pathophysiology and management of dengue hemorrhagic fever. Transfusion Alternatives in Transfusion Medicine 2006; 8: 3-11. https://doi.org/10.1111/j.1778-428X.2006.00025.x
- Mazzon M, Jones M, Davidson A, Chain B and Jacobs M: Dengue virus NS5 inhibits interferon-α signaling by blocking signal transducer and activator of transcription 2 phosphorylation. The Journal of Infectious Diseases 2009; 200(8): 1261-70.doi.org/10.1086/605847
- Guzman A and Istúriz RE: Update on the global spread of dengue. International Journal of Antimicrobial Agents. 2010; 36: 40-2.doi.org/10.1016/j.ijantimicag.2010.06.018
- Kellstein D and Fernandes L: Symptomatic treatment of dengue: should the NSAID contraindication be reconsidered?. Postgraduate Medicine 2019; 131(2): 109-16. doi.org/10.1080/00325481.2019.1561916
- Shen TJ, Hanh VT, Nguyen TQ, Jhan MK, Ho MR and Lin CF: Repurposing the antiemetic metoclopramide as an antiviral against dengue virus infection in neuronal cells. Frontiers in Cellular and Infection Microbiology 2021; 10: 606743.doi.org/10.3389/fcimb.2020.606743
- Bawaskar RS and Shinde VH: A Review of homoeopathic research in the prevention and treatment of dengue fever. Homœopathic Links 2019; 32(01): 010-7. DOI: 10.1055/s-0039-1687897
- Malavige GN, Jeewandara C, Wijewickrama A, Gunasinghe D, Mahapatuna SD, Gangani C, Vimalachandran V, Jayarathna G, Perera Y, Wanigatunga C and Dissanayake H: Efficacy of rupatadine in reducing the incidence of dengue haemorrhagic fever in patients with acute dengue: A randomised, double blind, placebo-controlled trial. PLoS Neglected Tropical Diseases 2022; 16(6): 0010123.doi.org/10.1371/journal.pntd.0010123
- Borges MC, Castro LA and Fonseca BA: Chloroquine use improves dengue-related symptoms. Memórias do Instituto Oswaldo Cruz 2013; 108(5): 596-9.doi.org/10.1590/S0074-02762013000500010
- Pambhar V, Mathur N, Mehta A, Mathur M, Mangalia R, Verma A and Patyal A: Effect of doxycycline and doxycycline with carica papaya on thrombocytopenia and leucopenia in acute dengue fever patients. Journal of Family Medicine and Primary Care 2022; 11(6): 3270-5.DOI: 10.4103/jfmpc.jfmpc_53_22
- Anekthananon T, Pukritayakamee S, Ratanasuwan W, Jittamala P, Werarak P, Charunwatthana P, Suwanagool S, Lawpoolsri S, Stepniewska K, Sapchookul P and Puthavathana P: Oseltamivir and inhaled zanamivir as influenza prophylaxis in Thai health workers: a randomized, double-blind, placebo-controlled safety trial over 16 weeks. Journal of Antimicrobial Chemotherapy 2013; 68(3): 697-707.doi.org/10.1093/jac/dks418
- Ooi EE: Repurposing ivermectin as an anti-dengue drug. Clinical Infectious Diseases 2021; 72(10): 594-5.https://doi.org/10.1093/cid/ciaa1341
- Xu TL, Han Y, Liu W, Pang XY, Zheng B, Zhang Y and Zhou XN: Antivirus effectiveness of ivermectin on dengue virus type 2 in Aedes albopictus. PLoS Neglected Tropical Diseases 2018; 12(11): 0006934.
- Lee IK, Lee WH, Yang KD and Liu JW: Comparison of the effects of oral hydration and intravenous fluid replacement in adult patients with non-shock dengue hemorrhagic fever in Taiwan. Transactions of the Royal Society of Tropical Medicine and Hygiene 2010; 104(8): 541-5.doi.org/10.1016/j.trstmh.2010.05.003
- Gratz CM: Citrate with procaine in fracture treatment. Journal of the American Medical Association 1943; 123(14): 927-.doi:10.1001/jama.1943.02840490055025
- Hung NT: Fluid management for dengue in children. Paediatrics and International Child Health 2012; 32(1): 39-42.doi.org/10.1179/2046904712Z.00000000051
- Unissa R, Jyothirmayi B, Mounica A, Tejaswini L, Latha GA, Nishitha G and Swati C: Dengue. Asian Journal of Research in Pharmaceutical Science 2018; 8(4): 192-202. doi.org/10.5958/2231-5659.2018.00033.4
- Mugade M, Patil SD, Kadam A, Ravariya K, Patel S and Shinde A: A Comprehensive Review on Fruit-Based Interventions in Dengue Treatment. doi.org/10.55248/gengpi.5.0524.1342
- Ashok AD, Kayalvizhi K and Ravivarman J: Indian vegetable diet in immunity development against viral diseases. Int J Curr Microbiol App Sci 2020; 9: 3411-7.doi.org/10.20546/ijcmas.2020.906.404
- Banerjee S, Sharma U and Kaur VP: Analysis of essential hematological parameters and nutrient levels of dengue patients aiming for quick recovery and sustainable immunity. doi.org/10.3390/molecules27092760
- Choudhary NL, Kumawat P, Patel P and Chishty N: Dengue and natural remedies for its prevention and control: a review. doi.org/10.22271/23487941.2022.v9.i6b.646
- Mugade M, Patil SD, Kadam A, Ravariya K, Patel S and Shinde A: A Comprehensive Review on Fruit-Based Interventions in Dengue Treatment doi.org/10.55248/gengpi.5.0524.1342
- Ashok AD, Kayalvizhi K and Ravivarman J: Indian vegetable diet in immunity development against viral diseases. Int J Curr Microbiol App Sc 2020; 9: 3411-7. doi.org/10.20546/ijcmas.2020.906.404
- Ahmed S, Finkelstein JL, Stewart AM, Kenneth J, Polhemus ME, Endy TP, Cardenas W and Mehta S: Micronutrients and dengue. The American Journal of Tropical Medicine and Hygiene 2014; 91(5): 1049.doi.org/10.4269/ajtmh.14-0142
- Saha D, Mandal S, Roy S, Mahanty S, Sinha NK, Maiti R and Majumder S: Micronutrients and Dengue Fever: Exploring their Role in Effective Management Strategies. DOI: 10.2174/0115734013340202241108062720
- Dussupt V, Modjarrad K and Krebs SJ: Landscape of monoclonal antibodies targeting zika and dengue: therapeutic solutions and critical insights for vaccine development. Frontiers in Immunology 2021; 11: 621043.doi.org/10.3389/fimmu.2020.621043
- Ormundo LF, Barreto CT and Tsuruta LR: Development of therapeutic monoclonal antibodies for emerging arbovirus infections. Viruses 2023; 15(11): 2177.doi.org/10.3390/v15112177
- Chan KR, Ong EZ and Ooi EE: Therapeutic antibodies as a treatment option for dengue fever. Expert Review of Anti-Infective Therapy 2013; 11(11): 1147-57.doi.org/10.1586/14787210.2013.839941
- Hassan M, Hassan A, Farooq M, Afzal S, Khan MA, Amin I, Shahid M, Idrees M and Shahid AA: Dengue vaccines: ongoing challenges and current status in the advancement of different candidates. Critical Reviews™ in Eukaryotic Gene Expression 2021; 31(5). DOI: 10.1615/CritRevEukaryotGeneExpr.2021038839
- Thisyakorn U and Thisyakorn C: Latest developments and future directions in dengue vaccines. Therapeutic Advances in Vaccines 2014; 2(1): 3-9.doi.org/10.1177/2051013613507862
- Prompetchara E, Ketloy C, Thomas SJ and Ruxrungtham K: Dengue vaccine: Global development update. Asian Pac J Allergy Immunol 2020; 38(3): 178-85.DOI 10.12932/AP-100518-0309
- Gholap AD, Khuspe PR, Pardeshi SR, Uddin MJ, Das U, Hatvate NT, Rojekar S, Giram P, Khalid M, Choonara YE and Faiyazuddin M: Achieving Optimal Health With Host‐Directed Therapies (HDTs) in Infectious Diseases A New Horizon. Advanced Therapeutics 2024; 2400169. doi.org/10.1002/adtp.202400169
- Nielsen DG: The relationship of interacting immunological components in dengue pathogenesis. Virology journal. 2009; 6: 1-7.doi:10.1186/1743-422X-6-211
- Liu T, Yang WQ, Xie YG, Liu PW, Xie LH, Lin F, Li CY, Gu JB, Wu K, Yan GY and Chen XG: Construction of an efficient genomic editing system with CRISPR/Cas9 in the vector mosquito Aedes albopictus. Insect Science 2019; 26(6): 1045-54.doi.org/10.1111/1744-7917.12645
- Liu X, Zhou X, Xie X and Li C: Advances in CRISPR/Cas9-Based Gene Editing Technology in Mosquitoes. Zoonoses 2024; 4(1): 971. DOI: 10.15212/ZOONOSES-2024-0020
- Lye DC, Archuleta S, Syed-Omar SF, Low JG, Oh HM, Wei Y, Fisher D, Ponnampalavanar SS, Wijaya L, Lee LK and Ooi EE: 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.
- Goh ZJ, Li R, Wang MX, Chia PY and Lim JT: Transfusion of blood products and clinical outcomes for patients with dengue fever: a systematic review and meta-analysis. In Open Forum Infectious Diseases 2024; 11(9): 507. US: Oxford University Press.doi.org/10.1093/ofid/ofae507
How to cite this article:
Ganguly G, Sahoo T, Ghosh S, Gupta A, Nayak A, Mojamel SK and Sahabuddin M: Emerging interventions & therapies for effective dengue management. Int J Pharmacognosy 2025; 12(2): 37-57. doi link: http://dx.doi.org/10.13040/IJPSR.0975-8232.IJP.12(2).37-57.
This Journal licensed under a Creative Commons Attribution-Non-commercial-Share Alike 3.0 Unported License.
Article Information
1
37-57
907 KB
72
English
IJP
Gourav Ganguly *, Tarpan Sahoo, Shibam Ghosh, Abhik Gupta, Arabinda Nayak, S. K. Mojamel and Mirza Sahabuddin
Department of Pharmacology, Bengal College of Pharmaceutical Technology, Dubrajpur, Birbhum, West Bengal, India.
kujhi.2008@gmail.com
27 January 2025
26 February 2025
27 February 2025
10.13040/IJPSR.0975-8232.IJP.12(2).37-57
28 February 2025