Skip to main content

Exploring plant-based dengue therapeutics: from laboratory to clinic

Abstract

Dengue virus (DENV) is a mosquito-borne virus that causes dengue fever, a significant public health concern in many tropical and subtropical regions. Dengue is endemic in more than 100 countries, primarily in tropical and subtropical regions of the world. Each year, up to 400 million people get infected with dengue. Approximately 100 million people get sick from infection, and 40,000 die from severe dengue. Unfortunately, dengue vaccine development is also marred with various complicating factors, as the forefront candidate vaccine performed unsatisfactorily. Moreover, the only licensed vaccine (Dengvaxia) for children 9 through 16 years of age is available in just a few countries. The treatment difficulties are compounded by the absence of an effective antiviral agent. Exploring plant-based therapeutics for dengue from the laboratory to clinical application involves a multi-stage process, encompassing various scientific disciplines. Individual investigators have screened a wide range of plant extracts or compounds for potential antiviral activity against DENV. In vitro studies help identify candidates that exhibit inhibitory effects on viral replication. Some of the most promising medicinal plants showing in vitro activity against DENV include Andrographis paniculate, Acorus calamus, and Cladogynos orientalis. Further laboratory studies, both in vitro and in animal models (in vivo), elucidate the mechanisms of action by which the identified compounds exert antiviral effects. Medicinal plants such as Carica papaya, Cissampelos pareira, and Ipomea batata exhibited potent platelet-enhancing activities while Azadirachta indica and Curcuma longa showed promising effects in both in vitro and in vivo studies. Based on positive preclinical results, researchers design clinical trials. This involves careful planning of trial phases, patient recruitment criteria, ethical considerations, and endpoints. The most important medicinal plants showing efficacy and safety in clinical trials include Carica papaya and Cissampelos pareira. This review suggests that several promising medicinal plants exist that have the potential to be turned into clinical drugs to treat dengue infection. However, in addition to developing synthetic and plant-based therapies against dengue infection, vector management strategies should be made robust, emphasizing the need to focus on reducing disease incidence.

Introduction

Viruses put an enormous burden on humanity and are the single most important source of infectious disease morbidity and mortality throughout the world [7, 31, 61]. Dengue virus (DENV) is a member of the Flaviviridae family and the flavivirus genus with four genetically related but different serotypes, DENV 1, 2, 3, and 4. Any one of the four DENV serotypes (DENV-1 to DENV-4) can cause an infection. The symptoms of dengue shock syndrome (DSS), which can result in major hemorrhage and shock and carry a 20% mortality risk if left untreated, are similar to those of dengue fever, including fever, nausea, vomiting, rash, and body pains [44]. DENV is spread by mosquitoes and affects millions of people each year. It is endemic in more than 100 countries, primarily in tropical and subtropical regions of the world. Almost 7.6 million dengue cases were reported to the World Health Organization (WHO), including 3.4 million confirmed cases with 16,000 severe cases, and over 3000 deaths. A notable rise has been observed in the Americas, where the number of cases crossed 7 million by the end of April 2024, as compared to the annual high of 4.6 million cases in 2023. Additionally, vector control and surveillance cost approximately 500 million dollars [107, 123].

Aedes aegypti, the primary vector, is a daytime mosquito that may infect multiple victims at once, and nests in artificial water-collection containers. Aedes albopictus, a less efficient vector, is expanding its geographic range into temperate and tropical areas. The main reasons for the development of Aedes mosquitoes and the increased incidence of dengue are population growth, high population density, rural-to-urban migration, deteriorated urban environments, lack of dependable piped water, and inadequate mosquito control programs [104]. The mobility of humans has risen as a result of modern transportation, which has contributed to the rapid spread of dengue viruses around the world [45]. The likelihood of dengue outbreaks in temperate zones is raised by both global warming and the spread of Aedes mosquitoes [128].

Vaccine development for diseases like dengue faces significant challenges, including the issue of Antibody-Dependent Enhancement (ADE), where antibodies from a previous infection may worsen a subsequent infection with a different virus type [117]. As far as antiviral agents are concerned, direct-acting antivirals (DAAs) are a promising alternative, but they are prone to resistance development. Host-directed antivirals (HDAs) offer another potential strategy by interfering with cellular pathways to disrupt the virus's replication environment. However, a narrower therapeutic window and interference with normal physiological function make it a less desirable candidate [117]. The alarming rise in dengue cases and glaring treatment gaps for its management are compelling researchers to look for plant-based therapeutic agents. In this review, many promising plant-derived treatments have been mentioned, like Andrographis paniculate, Carica papaya, and Curcuma longa [131]. These studies should be scaled up and replicated to combat the burgeoning threat of dengue, as climate change is only going to exacerbate this menace [12].

Epidemiology of dengue infection

Aedes mosquitoes are responsible for the transmission of dengue, a disease that affects over 3 billion people worldwide and is particularly prevalent in tropical and subtropical areas [132]. As of 30 April 2024, over 7.6 million dengue cases were reported to the WHO, including 3.4 million confirmed cases. Moreover, 16,000 severe cases and over 3000 deaths have also been reported. A notable rise in dengue cases has been observed at the global level in the last half a decade, particularly in the Americas, where the number of cases had already exceeded 7 million by the end of April 2024, going beyond the annual high of 4.6 million cases in 2023. Currently, 90 countries have known active dengue transmission in 2024, not all of which have been captured in formal reporting [123] affecting 5 billion people, and some places have mortality rates are extremely high. More than 100 countries are affected by dengue infections, including the United States, and Europe [92, 125]. In Pakistan, the incidence of dengue has risen dramatically over the past twenty years [10, 52, 137] becoming a serious public health issue due to high mortality, morbidity, and loss of productivity. The country has frequently reported severe dengue outbreaks, including in 1994, 1995, 1997, 2006, 2007, 2011, and 2019 [75]. The climate conditions of Pakistan provide a favorable breeding ground for DENV replication and transmission. The loss of human capital linked with the aftermath of the dengue epidemic is growing. 16,580 confirmed dengue cases, with 257 deaths, were registered in 2010. Whereas in 2019, 47,000 dengue cases and 75 deaths were recently reported [75]. Until November 2021, the country recorded a total of 24,146 dengue cases and 183 associated deaths [122]. Notably, the record-breaking rainfall in August of 2022 and subsequent floods catalyzed the increase in dengue cases in October 2022 [54, 113].

DENV genome, its proteins & their role in dengue infection

It is proposed that the cell and tissue tropism of DENV could profoundly influence the prognosis of DENV infections. In vitro, and autopsy studies indicate the critical role played by three organ systems in dengue pathogenesis: the liver, the immune system, and endothelial cell (EC) linings of blood vessels [74]. DENV has a single-strand positive-sense RNA genome of 11 kb (Fig. 1). The genome encodes a polyprotein that is translated into seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) and three structural proteins (capsid protein—CP, envelope protein—EP, membrane protein- MP). Non-structural proteins play vital roles in virus entry, replication, assembly, and pathogenesis. Moreover, non-structural proteins regulate DENV replication, influence immunological responses, induce vascular leakage, and assist in viral RNA synthesis and polyprotein cleavage. [60]. According to [19] Dengue severity arises from a complex interplay involving the virus, host genes (HLA, and IL-10), and immune responses (ADE, memory T cells).

Fig. 1
figure 1

Dengue Virus Structure and Genomic Processing. An overview of the Dengue Virus (DENV), showing its structural anatomy with the RNA genome, capsid, envelope, and E & M proteins. It outlines the positive-sense RNA genome's division into regions coding for structural and non-structural proteins and details the translation and processing of these proteins from a polyprotein precursor into individual components crucial for the virus's life cycle

DENV is presumably injected following the mosquito bite into the epidermis and dermis of an organism. This results in the infection of immature Langerhans cells, also known as epidermal dendritic cells (DC) and keratinocytes [71, 129]. Moreover, DENV was found to not only activate the DCs but also disrupt their physiological dermal migration process [71]. Afterward, the infected cells leave the site of infection to the lymph nodes, initiating the recruitment of monocytes and macrophages, which fall prey to the infection. This leads to the spread of infection to viral dissemination through the lymphatic system. This primary surge in viremia infects several cells of the mononuclear lineage, including blood-derived monocytes [35], myeloid DC [24], and liver and splenic macrophages [23, 47]. It is noteworthy that during secondary infections with heterologous DENV, high concentrations of DENV-specific immunoglobulin G (IgG) form a complex newly produced virus, which adheres to and is subsequently taken up by the mononuclear cells [74]. Following infection, mononuclear cells predominantly die by apoptosis [36]. Abortively infected or bystander-DC releases several mediators that are involved in inflammatory [73] and hemostatic [48] responses of the host. In order to undermine the integrity of the endothelial cell monolayer in blood vessels, the DENV NS1 protein interacts with Toll-Like Receptors 4 (TLR4) activating macrophages and peripheral blood mononuclear cells (PBMCs). Apoptotic pathways are triggered and platelets are destroyed when NS1 proteins from cells infected with DENV interact with TLR4 on the platelets' plasma membrane. This happens because P-selectin is expressed more often [30]. Furthermore, stromal cells of bone marrow also demonstrated a susceptibility to DENV infection [82]. Some of the important players involved in dengue infection are outlined in Fig. 2.

Fig. 2
figure 2

Pathophysiology of Dengue-Induced Inflammation and Shock. The pathophysiological mechanisms triggered by Dengue virus infection that lead to inflammation and potentially to cytokine storm and shock. Upon infection, the virus activates platelets and monocytes, leading to the release of inflammatory cytokines (IL-6, IL-10, IL-12, IL-18) and TNF-α. Macrophages produce nitric oxide, while mast cells release VEGF and secretory Phospholipase A2 (sPLA2). These mediators contribute to endothelial activation and NF-κB production, further promoting inflammation. The combined effects result in increased vascular permeability (plasma leakage), contributing to the development of shock

Important anti-dengue drug targets

Broadly, anti-viral targets for dengue treatment are classified as viral entry inhibitors, fusion inhibitors, capsid (C) protein storage inhibitors, viral assembly inhibitors, and viral maturation inhibitors [2]. Several studies have identified both structural and nonstructural proteins as important drug targets including the structural proteins (C-protein, E-protein, prM), as well as nonstructural proteins (NS1, NS2a, NS2b, NS3, NS4a, NS4b, and NS5) [86].

Targeting structural proteins, the building blocks of viruses enervates and eventually kills the virus. Therefore, they can be promising drug targets for treating DENV infections [9]. Notably, E-protein has been the major target for vaccine development owing to its vital role in virus attachment and penetration into host cells [13, 70]. Another important drug target is prM. It is believed to be a chaperon for E-protein to ensure its conformational changes during different steps of the secretory pathway and to produce infectious viral particles, particularly blocking membrane fusion [136].

C-protein, located in the cytosol, is the first protein translated by the viral genome followed by prM. However, due to the least conservation of C-protein across DENV serotypes, this target is less favored [83, 103].

Non-structural NS1 is an enigmatic multifunctional, dengue glycoprotein. Due to the high conservation of NS1 among all DENV serotypes it is a favorable target for drug and vaccine development [80, 98]. Similarly, NS3 the extensively studied, multifactorial protein is among the most conserved DENV proteins. Notably, 77% of amino acids of NS3 are similar in all four serotypes, making it a potential target for drug and vaccine development [18]. NS4B, which is involved in host membrane modification, and essential for replication, emerges as one of the most promising drug targets as novel inhibitors targeting NS4B and its interaction network are entering clinical studies [68, 130]. Similarly, NS5 remains a crucial protein for DENV replication and pathogenesis. Targeting NS5 offers a strategic avenue for designing effective antiviral drugs [83].

Current treatments and associated challenges

The therapeutic vacuum in treating dengue infection is being filled by symptomatic treatment and fluid resuscitation methods due to the absence of a potent anti-dengue medicine. This makes dengue management more challenging. At the moment, vector control is the sole method for preventing disease, despite a poor cost–benefit ratio. Supportive care, including antipyretics, analgesics, and bed rest is the basic treatment for dengue patients. Urgent resuscitation with intravenous fluids is necessary for DSS patients, with Ringer's lactate being effective for mildly serious dengue and starch or dextran being preferred for more critical cases [96]. Severe dengue, characterized by plasma leakage, severe bleeding, or organ impairment, entails significant morbidity and mortality if not treated timely. Multi-organ impact of dengue can lead to problems like fluid overload and poor metabolism of analgesics, especially for patients with comorbidities [119].

The most commonly used antipyretic and analgesic is acetaminophen, which comes with its own set of challenges like liver and kidney toxicity [39]. Inhibitors of cyclooxygenase 2 may be helpful as anti-inflammatory and antioxidants, but they may also inhibit platelets [8, 15]. Moreover, ribavirin has been effective in treating a number of RNA viruses, however, it has not been successful in treating DENV, and it has a cytostatic impact on DENV-infected cells. Similarly, a promising alternative therapy called RNA interference (RNAi) destroys unnecessary genetic material to shield the host from viral infections. Notably, the RNAi machinery can boost HCV replication, and flavivirus RNA is resistant to RNAi [65]. Furthermore, prophylactic platelet transfusion is prohibited and is reserved for managing severe cases. In addition to the high costs of platelet transfusion, blood-safety concerns create more complications. Numerous studies have documented transfusion-transmitted dengue, with the first cases being reported in China in 2002 and Singapore in 2008 [78].

Dengue vaccine development is also marred by various complicating factors. Unfortunately, the forefront candidate vaccine performed unsatisfactorily, and its efficacy depends on serostatus and age factors. The observations from clinical studies depicted ambiguity regarding the efficacy of the dengue vaccine [50]. Moreover, the only licensed vaccine (Dengvaxia) for children 9 through 16 years of age is available in just a few countries. Despite its licensure for clinical use, the vaccine has performed below par efficacy in children and dengue-naive individuals and notably resulted in an increased risk of developing severe dengue in young, vaccinated recipients [94, 115]. In addition to absence of a suitable animal model, another major hurdle in vaccine development is ADE. In the case of ADE, antibodies generated after the first infection can actually facilitate the entry of the virus into cells when a person is infected with a second, different type of DENV, making the infection more severe [58]. ADE enhances DENV NS1 production, an endothelial toxin, increasing vascular permeability that leads to severe, even fatal, dengue [40, 43].

Direct-acting antivirals (DAA), which interact with viral proteins to exert antiviral function, offer a promising approach with lower toxicity and a broad therapeutic window. However, a downside of DAA is a higher risk for viral resistance [117]. The most extensively studied structural protein and non-structural antiviral drug targets are the E protein, and NS5 and NS3, respectively. Despite numerous DAA identified, a few are validated through in vivo studies. Furthermore, only one DAA, balapiravir, has been tested in clinical trials but no remarkable difference in cytokine profile, plasma viral load, and fever clearance time between the placebo and balapiravir groups was reported [25].

Another potential approach is interference with cellular pathway to prevent viral commandeering of cellular machinery to create a favourable environment for its replication. The compounds capable of such intervention are called as host-directed antivirals (HDA). HDA not only presents a promising avenue for broad-spectrum treatment but also has a reduced risk of resistance development [59]. In comparison to DAA, HDA typically have a narrow therapeutic and toxicity window, as they may also interfere with cell homeostasis[4]. The most studied cellular target is α-glucosidase, which facilitates proper protein folding and maturation. One of the Few studied HDA in clinical trials is the α-glucosidase inhibitor UV-4B. This drug exhibited a good safety profile up to a concentration of 1000 mg [85]. However, a follow-up study regarding the pharmacokinetics of UV-4B in healthy individuals was terminated.

Furthermore, a live-attenuated tetravalent DENV vaccine has been developed by Takeda (TAK-003). The vaccine is comprised of a DENV-2 backbone and 3 chimeric viruses with pre-membrane and envelope protein genes from three DENV strains, DENV 1, 3, and 4 [88]. Notably, the vaccine is getting licensed for use around the world, like in Brazil, Argentina, Indonesia, Colombia, Thailand, the European Union, and the United Kingdom. Clinical trials have demonstrated, that upon giving 2 subcutaneous injections 3 months apart, TAK-003 is well-tolerated and immunogenic not only in healthy adults from dengue-nonendemic areas but also in adults and children in dengue-endemic regions [20, 21]. However, in Takeda's February 2024 4–5-year QDENGA efficacy report, out of 3174 patients vaccinated and seronegative aged 4–16 years, only 11 were hospitalized with DENV 3 infections, while from 1832 seronegative patients on placebos, only 3 were hospitalized, giving a negative efficacy of 11·6%, similar to that of Dengvaxia [43, 110]. Moreover, overall vaccine efficacy (VE) was 80.2% against virologically confirmed dengue (VCD) caused by any serotype from 30 days after the second dose to the end of year 1 [22]. The overall VE against hospitalized dengue patients from 30 days after the second dose to the end of 18 months was 90.4% [20]. At 4.5 years postvaccination mark, the cumulative VE from the first dose against VCD was 61.2%, while 53.5% in baseline seronegative participants against all 4 dengue serotypes, and 64.2% in baseline seropositive subjects for any dengue serotype [116]. The evaluation study suggested that irrespective of the baseline serostatus, the vaccine is efficacious against DENV 1 and 2. On the other hand, VE against DENV 3 and 4 was only evident in baseline seropositive subjects. As for baseline seronegative recruits, data suggested no satisfactory efficacy against DENV 3, due to the low number of DENV 4 cases a proper assessment of VE could not be made [116]. The Strategic Advisory Group of Experts on Immunization recently recommended the use of QDENGA in dengue virus-endemic countries in settings of high transmission, for children aged 6–16 years irrespective of serostatus [124]. The approval was given in light of studies reporting an absence of a statistically significant number of severe DENV 3 infections in QDENGA vaccinated seronagative patients [124].

Medicinal plants as remedies for dengue infection

The use of medicinal plants in treating dengue infections is an area of growing interest, especially given the limited availability of specific antiviral treatments for dengue. Medicinal plants have been used for centuries to treat various human illnesses [5, 49, 53, 102]. Various plants have been studied for their potential to alleviate symptoms or affect the virus directly. Some plants have been found to possess antiviral properties that may be effective against the dengue virus. These plants can inhibit the replication of the virus or interfere with its ability to enter host cells. Certain plants are known for their ability to alleviate symptoms associated with dengue, such as fever, pain, and inflammation. They may possess analgesic, antipyretic, or anti-inflammatory properties [42, 99]. Some medicinal plants can strengthen the immune system, helping the body to fight off the virus more effectively. In many cultures, specific plants have been used traditionally to treat symptoms similar to those of dengue fever. These traditional uses often guide scientific research into the potential of these plants as dengue remedies.

Rural populations around the world employ a variety of herbs from the Lamiaceae family to cure DENV infections. As shown in Table 1, other widely used species include Carica papaya (Caricaceae) and Euphorbia hirta (Euphorbiaceae). Several plant components are employed, including the leaves, roots, stems, and flowers, in decoctions, infusions, and leaf juice for treating dengue infection. In this section, we describe and discuss medicinal plants with in vitro and in vivo activities against DENV. We also discuss clinical trials in which medicinal plants were employed for treating dengue infection.

Table 1 In vitro studies for pharmacologically active medicinal plants DENV infection treatment

Medicinal plants with in vitro activity

In vitro screening of drugs for dengue treatment plays a crucial role in the early stages of drug development. In vitro screening allows researchers to test a wide range of compounds for their antiviral activity against the dengue virus. This initial screening helps identify potential drug candidates that exhibit inhibitory effects on viral replication. In this section, we describe and discuss some of the most important medicinal plants showing promising anti-dengue potential as tested in vitro (Table 1 and Fig. 3).

Fig. 3
figure 3

Several medicinal plants have shown activities against various DENV targets. Extracts of plants or phytocompounds isolated from them were effective asNS1 inhibitor (Quersus lucitanica), NS5 inhibitor (Acorus calamus), vRNA inhibitor (Houttuynia cordata), and viral entry inhibitor (Orthosiphon stamineus)

Alternanthera philoxeroides

Alternanthera philoxeroides, commonly known as alligator weed, belongs to the Amaranthaceae family. Various polarity-based extracts of the whole plant were assessed using C6/36 cell lines. Cytotoxicity was only exhibited by the concentrations exceeding 320 μg/ml. All four extracts demonstrated inhibitory effects on the dengue virus, however, petroleum ether extracts exhibited the strongest inhibitory effects on the dengue virus (ED50 = 47.43) among the four extracts [57].

Andrographis paniculata

Andrographis paniculata, commonly known as the King of Bitters, belongs to the family Acanthaceae. Ethanolic and aqueous extracts of A. paniculata at a concentration above 500 μg were toxic to Vero cells; also, they exhibited antiviral effects, with 55–97% cell viability recorded in dengue-infected cells. The study determined the maximal non-toxic dose (MNTD) and highlighted the potential of A. paniculata as an anti-dengue agent, showing 75% viral inhibition [97].

Acorus calamus

Methanolic extract of Acorus calamus (Acoraceae), prepared from A. calamus leaves, was subjected to in vitro laboratory experiments combined with in-silico analysis. The results indicated that the extract was non-toxic to Huh7it-1 cells while demonstrating a significant inhibition of DENV-2. Molecular docking analysis revealed that artesunic acid in A. calamus is potential inhibitor of the NS5 protein of the dengue virus [101].

Cladogynos orientalis

Cladogynos orientalis (Family Rubiaceae) exhibited significant inhibitory activity against DENV-2 in Vero cells. Ethanol extracts C. orientalis showed 34.85% inhibitory activity, at a concentration of 12.5 µg/ml. Additionally, C.orientalis, exhibited the ability to inactivate viral particles by 52.9% at a concentration of 100 µg/ml [62].

Euphorbia hirta

Euphorbia hirta (Euphorbiaceae) is traditionally used in the Philippines to combat dengue virus. The ethyl acetate layer of the plant extract exhibited the most potent antiviral activity against dengue virus serotypes 1 and 2. It also significantly reduced the plaque-forming capacity of the dengue virus, suggesting potential antiviral properties [114].

Flagellaria indica

The ethanol extract of Flagellaria indica (Flagellariaceae) at a concentration of 12.5 µg/ml showed 45.52% inhibitory effects on DENV-2 replication. However, the identity of the active constituents and underlying mechanism was not explored by the investigators [62].

Flacourtia ramontchi

Flacourtia ramontchi, colloquially known as ramontchi, governor's plum, and Indian plum, belongs to the Salicaceae family. The stem bark extract of F. ramontchi exhibited significant anti-dengue activity. Bioassay-guided isolation led to the identification of six new phenolic glycosides, including flacourtosides A–F (1–6), scolochinenoside D, and betulinic acid 3β-caffeate. It was observed that betulinic acid 3β-caffeate, flacourtosides A and E (1 and 5), and scolochinenoside D exhibited significant inhibition in the DENV RNA polymerase assay. Notably, betulinic acid 3β-caffeate demonstrated an IC50 of 0.85 ± 0.1 μM, highlighting its potential as a candidate for further exploration as an anti-dengue therapeutic agent [26].

Faramea bahinensis

A methanol extract from the leaves of Faramea bahiensis, a plant belonging to the Rubiaceae family, demonstrated in vitro non-cytotoxicity and significant anti-DENV-2 activity in human hepatocarcinoma (HepG2) cell lines. Further fractionation led to the isolation of a new flavanone glycoside, 5-hydroxy-4′-methoxy-flavanone-7-O-ß-d-apiofuranosyl-(1 → 6)-ß-d-glucopyranoside, in addition to known flavanone glycosides. The treatment of DENV-2-infected HepG2 cells with the new flavanone glycoside resulted in the control of viral replication and subsequent reduction in the number of infected cells (12%), infectious particles in the culture supernatant (97%), and the number of DENV-2 RNA copies in HepG2 cells (67%) [84].

Houttuynia cordata

An aqueous extract of aerial stems and leaves of Houttuynia cordata exhibited inhibitory effects on intracellular DENV-2 RNA production and dengue protein expression in HepG2 cells. The extract exhibited a protective effect on virion release from the infected monkey kidney cell line (LLC-MK2), with hyperoside identified as the predominant bioactive compound. Importantly, the extract was found to be non-genotoxic, further supporting its potential safety for consumers [66]. An ethanolic extract of Houttuynia cordata was obtained by soaking the herb in 80% ethanol, followed by filtration, concentration, and freeze-drying. The ethyl acetate (EA) fraction of H. cordata, particularly quercetin exhibited significant anti-DENV-2 activity, while the combination of quercetin and quercitrin showed a synergistic effect without inducing cytotoxicity. The acute oral toxicity assessment of the EA fraction in mice revealed no signs of toxicity or adverse effects [33].

Faramea hyacinthina and Faramea truncate

The in-vitro assays employed the defatted fractions of methanol extracts from the leaves of Faramea hyacinthina and Faramea truncate (family Rubiaceae) exhibited non-cytotoxicity and demonstrated anti-DENV2 activity in HepG2 cells. Several antiviral compounds were isolated including the antiviral flavanone (2S)-isosakuranetin-7-O-β-d-apiofuranosyl-(1 → 6)-β-d-glucopyranoside (1), diastereoisomeric epimers (2S) + (2R) of 5,3′,5′-trihydroxyflavanone-7-O-β-d-apiofuranosyl-(1 → 6)-β-d-glucopyranoside (2a/2b), narigenin-7-O-β-d-apiofuranosyl-(1 → 6)-β-d-glucopyranoside (3), rutin (4), quercetin-4′-β-d-O-glucopyranosyl-3-O-rutinoside (5), kaempferol-3-O-rutinoside (6), erythroxyloside A (7), and asperuloside (8). Compounds 4 – 8 were reported for the first time in Faramea species [17].

Justicia adhatoda

Aqueous extracts of Justicia adhatoda (family, Acanthaceae), were encapsulated in phytoniosomes (PN) and their antiviral activity against DENV-2 was evaluated. The PN formulations, particularly AGN4, showed significant inhibition of plaque formation in in-vitro conditions, suggesting the potential of J. adhatoda leaf extract in combating dengue virus. However, the mechanism of inhibition was not investigated [126].

Piper retrofractum

Several Thai medicinal plants, including Piper retrofractum belonging to the Piperaceae family, were investigated in in-vitro anti-dengue activity. In the study, dichloromethane and ethanol extracts from P. retrofractum demonstrated inhibitory activity against DENV-2 in Vero cells. At a concentration of 12.5 µg/ml, the ethanol extract exhibited a significant inhibitory activity of 53.53%. Additionally, at a concentration of 100 µg/ml, the ethanol extract of P. retrofractum showed an 84.93% inhibitory activity against DENV-2 viral particles. The cytotoxicity evaluation revealed a CC50 of 625 µg/ml for the ethanol extract of P. retrofractum, indicating its safety at this concentration. The mechanisms underlying its antiviral effects were not elucidated in the study [62].

Psidium guajava

An in vitro study model involving VERO cells was employed to assess the cytotoxicity and anti-DENV effects of ethanolic extracts from Psidium guajava, a plant belonging to the Myrtaceae family [118]. The most promising fraction, Pg-YP-I-22C, exhibited high. Further analysis revealed several phytocompounds, including catechin, gallic acid, hesperidin, naringin, and quercetin. In-vitro evaluations demonstrated significant antiviral effects, with catechin exhibiting 100% inhibition with a pre-treatment strategy and 91.8% with a post-treatment strategy. Molecular docking studies indicated favorable interactions, particularly with the viral envelope protein. The study sheds light on guava's potential as a source of antiviral compounds, with catechin emerging as a promising candidate for further investigation.

Phyllanthus amarus

An aqueous cocktail extract of Phyllanthus amarus (Phyllanthaceae family), excluding roots, was prepared, and processed into extracts using water, diethyldithiocarbamic acid, and formic acid. The study used in vitro experiments utilizing Vero cells as the model. Notable phytcompounds identified included gallic acid, geraniin, syringin, and corilagen. The MNTD on Vero cells was determined to be 250 μg/ml. Moreover, in vitro antiviral experiments against DENV2 revealed that Phyllanthus amarus exhibited significant activity, particularly in simultaneous treatment, resulting in an 83–95% reduction of virus inhibition. Proteomic analysis revealed altered expression of various host and viral proteins involved in crucial processes such as viral entry, replication, and cellular metabolism, suggesting Phyllanthus amarus as an important source of inhibitors against dengue virus [67].

Quersus lucitanica

Quersus lusitanica, commonly known as the Portuguese oak, belongs to the Fagaceae family. A crude extract derived from Q. lusitanica seeds investigated against DENV-2 using C6/36 cells, a cloned cell line derived from Aedes albopictus larvae, revealed dose-dependent antiviral activity. At its MNTD of 0.25 mg/ml, the Q. lusitanica extract completely inhibited various virus concentrations, demonstrating its effectiveness against cytopathic effects. Proteomic analysis showed the extract's impact on the NS1 protein expression, a glycoprotein crucial for flavivirus viability. The results exhibited down-regulation of NS1 protein expression in infected C6/36 cells upon treatment with Q. lusitanica extract. The promising inhibitory effects of Q. lusitanica extract on DENV-2 replication, showcase its potential as an antiviral agent in both conventional cell culture and proteomic analyses [79].

Rhizophora apiculata

Rhizophora apiculata is a mangrove plant belonging to the family Rhizophoraceae. An ethanol extract of R. apiculata plant exhibited a significant inhibitory activity against DENV-2 strain 16,681, in Vero cells with 56.14% inhibition at a concentration of 12.5 µg/ml. Moreover, R. apiculata extract demonstrated the ability to inactivate viral particles. The results suggest an anti-dengue potential of R. apiculata [62].

Spondias mombin & Spondias tuberosa

The plant extracts obtained from the leaves of two Spondias species of Anacardiaceae family, namely Spondias mombin and Spondias tuberosa, indigenous to Brazil, showed promising activity against DENV-2 evaluated in vitro using C6/36 cells, a cloned cell line derived from larvae of Aedes albopictus [109]. The study focused on the identification of phenolic compounds, including quercetin, rutin, and ellagic acid, through HPLC analysis. The extracts and compounds demonstrated non-toxicity to the cells at concentrations up to 1000 µg/mL. The antiviral activity revealed a significant inhibition of virus replication by rutin and quercetin at a concentration of 500 µg/mL. Notably, rutin exhibited a lower IC50 value (362.68 µg/mL) compared to quercetin (500 µg/mL), suggesting its greater potency. The study underscores the potential of Spondias species, particularly rutin and quercetin, as candidates for the development of anti-DENV agents.

Uncaria tomentosa

Uncaria tomentosa (family: Rubiaceae), commonly known as cat's claw, is a large woody vine native to the Amazon and Central American rainforests. Given the plant’s rich history of medical use, a hydro-alcoholic extract of U. tomentosa, as well as its pentacyclic oxindole alkaloid-enriched and non-alkaloid fractions were investigated utilizing human monocytes infected with DENV-2. Results indicated that both the extract and the alkaloidal fraction exhibited inhibitory activity against DENV-2, reducing the rates of DENV-2 antigen-positive cells. Additionally, the alkaloidal fraction indicated strong immunomodulation by decreasing levels of pro-inflammatory cytokines TNF-α and IFN-α, while modulating anti-inflammatory cytokine IL-10. These findings suggest that the pentacyclic oxindole alkaloids from U. tomentosa hold promise as potential candidates for clinical application in Dengue Fever treatment [100].

Annona muricata

An aqueous leaf extract of Annona muricata, commonly known as soursop, belonging to the family Annonaceae, was evaluated on Vero cells, revealing a CC50 of approximately 2.5 mg/mL and a 50% Effective Concentration (EC50) of about 0.20 mg/mL. Notably, the SI of the extract against DENV-2 was more than 10, indicating its potential as an antiviral agent. A. muricata demonstrated greater efficacy in inhibiting viral replication in post-treatment compared to pre-treatment. The phytochemical analysis identified over 200 chemical compounds, including alkaloids, phenols, and acetogenins. The study suggests that A. muricata aqueous leaves extract holds promise as a nature-based antiviral drug, particularly effective in the early stages of viral replication. However, the active compounds responsible for the observed effects were not identified in this study [120, 121].

Catharanthus roseus

Catharanthus roseus of the family Apocynaceae, commonly known as Madagascar periwinkle, is a medicinal plant with a rich history of traditional use for treating various diseases. A methanol extract from the leaves of C. roseus showed a CC50 of 0.13 mg/mL in vitro cytotoxicity tests using Vero cells. The antiviral activity against DENV-2 was also assessed through foci-forming unit reduction assays (FFURA), including post-treatment, pre-treatment, and virucidal tests. The results indicated that C. roseus extract exhibited a concentration-dependent antiviral effect, with more than 60% reduction in foci observed in the post-treatment assay at the highest concentration (0.078 mg/mL). The extract showed weak activity in pre-treatment and mild virucidal activity. The EC50 against DENV-2 was 0.025 mg/mL, resulting in a SI of 5.2 [1].

Orthosiphon stamineus

An aqueous leaf extract of Orthosiphon stamineus (Lamiaceae), commonly known as cat's whiskers or Java tea, revealed a CC50 value of 5 mg/ml, indicating non-cytotoxicity to the cells when tested against DENV-2 using Vero cells [120, 121]. The antiviral activity against DENV-2 was demonstrated through morphological changes and confirmed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, showing a selective index of 13. The study suggests that the extract from O. stamineus contains phytochemical compounds with non-cytotoxic and potential antiviral activities. This plant species holds promise as a candidate for further activity-monitored fractionation to identify active principles.

Hippophae rhamnoides

A leaf extract of Hippophae rhamnoides, of the Elaeagnaceae family, demonstrated a significant anti-dengue effect at a concentration of 50 μg/ml, comparable to Ribavirin, a commercially available anti-viral drug. The treatment with H. rhamnoides leaf extract led to a significant decrease in TNF-α and an increase in IFN-γ production in dengue-infected cells. Additionally, plaque assay results indicated a reduction in plaque numbers following the treatment, confirming its significant anti-dengue activity [55].

Faramea bahinensis, Faramea hyacinthina & Faramea truncate

An investigation centered on the methanolic extracts of three Faramea species – Faramea bahiensis, F. hyacinthina, and F. truncata, hailing from the Rubiaceae family revealed important anti-DENV2 activities. Utilizing online high-performance liquid chromatography-diode array detector-electrospray ionization tandem mass spectrometry (HPLC–DAD-ESI–MS/MS), the study identified 31 phenolic compounds, including flavonoid O- and C-glycosides, phenolic acids, and one coumarin. Notably, the stem extracts of F. hyacinthina and F. bahiensis exhibited significant anti-DENV2 activity comparable to their leaves. However, F. truncata demonstrated increased cytotoxicity [127]

Garcinia mangostana

Α-mangostin, derived from the fruit of G. mangostana (Guttiferae) [28], showed promising dengue treatment potential. Notably, at the concentrations of 20 and 25 μM, α-mangostin reduced DENV production by more than tenfold and 1,000-fold, respectively. Moreover, α-mangostin effectively inhibited the infection of immature monocyte derived dendritic cells (moDCs) by all four serotypes of DENV. α-mangostin, at 25 μM inhibited DENV at the early stage of replication. In addition, the phytocompound significantly reduced cytokine/chemokine (TNF-α, CCL4, CCL5, CXCL10, IL6, IL1β, IL10, and IFN-α) transcription in DENV-infected immature moDCs. This makes α-mangostin a suitable candidate for dengue treatment, as it demonstrates a distinct ability to thwart DENV-induced ‘cytokine storm’ [134].

Hystrix brachyuran

Traditionally, porcupine dates are used in dengue fever treatment. To study the anti-dengue compounds and their activity, two porcupine dates, black date and powdery date, from H. brachyuran (Hystricidae) were investigated. Methanol crude extracts were prepared from these dates to isolate tannin-rich fractions. The results revealed that all the extracts exhibited antiviral activity against DENV-2 in Vero cells. The IC50 of black date and powdery date tannin-rich fractions were 25 µg/mL and 11 µg/mL, respectively. Similarly, te fractions were highly virucidal (IC50 11 µg/mL) than methanol crude extracts with an IC50 value of 52–66 µg/mL. Notably, all the extracts inhibited the attachment of DENV-2 by at least 80% [95].

Medicinal plants with in vivo activity

In vivo screening of drugs for dengue treatment is a critical step in the drug development process, after the in vitro studies. In vivo studies involve testing potential drug candidates in living organisms, typically animal models, and provide valuable insights into the drug's efficacy, safety, and pharmacokinetics. The following plants showed potent in vivo activities in enhancing platelet, and thrombocyte count as well as attenuating the cytokine storm via downregulatio of pro-inflammatory cytokines.

Alternanthera sessillis

A study investigated the platelet augmentation activity of the leaf extract from Alternanthera sessilis (Amaranthaceae) to assess their potential as a therapy for thrombocytopenia. Platelet reduction was induced in Sprague–Dawley rats using anagrelide, and subsequent administration of plant extracts showed significant platelet augmentation activity. Notably, the percentage increase of mean platelet counts with A. sessilis was found to be 93.17% (p = 0.0001) [14].

Carica papaya

Carica papaya, commonly known as papaya, which belongs to the Caricaceae family (Table 2). An investigation used a novel approach involving a suspension of powdered C. papaya leaves in palm oil. Conducted in mice, the in vivo study administered 15 mg of powdered leaves/ kg body weight to different groups. The results demonstrated a significant increase in thrombocyte counts at multiple time points for the C. papaya leaf formulation group [105]. In a separate study, the authors aimed to evaluate the platelet augmentation activity of C. papaya extract in thrombocytopenic disorders. The in vivo study involved inducing platelet reduction in Sprague–Dawley rats and administering the plant extracts for nine days. C. papaya leaf extract demonstrated a notable increase in mean platelet counts (125.87%) [14].

Table 2 In vivo & in vivo/in vitro studies for pharmacologically active medicinal plants DENV infection treatment

Ipomea batatas

In another study, [14] investigated the platelet augmentation activity of Ipomoea batatas (Convolvulaceae), commonly known as sweet potato, in the context of thrombocytopenia. The study was conducted using Sprague–Dawley rats with induced platelet reduction through the administration of anagrelide. The aqueous extracts of I. batatas, particularly the violet and green varieties, demonstrated significant platelet augmentation activity, with a percentage increase of mean platelet counts by 106.07% and 107.88%, respectively. Further exploration is recommended to identify the specific phytochemical constituents responsible for this activity and to establish the safety profile of the plant extract.

Crocus sativus

C. sativus (Iridaceae) is renowned for immunomodulatory and anti-inflammatory properties. A study looking into C. sativus anti-dengue effects found that crocetin (C. sativus-derived natural compound) reduced DENV-induced apoptosis in DENV-infected mice at the dose of 50 mg/kg. Moreover, it significantly down regulated the pro-inflammatory cytokine expression, in addition to modulating antioxidant status in DENV-infected mice. Furthermore, crocetin demonstrated the ability to reduce the nuclear translocation of NF-kB. Another notable finding was that crocetin improved host responses that reduces liver injury in DENV-infected mice [111].

Medicinal plants in clinical trials

Clinical trials for drugs intended for dengue treatment are essential for several reasons, and their importance lies in advancing our understanding of the drug's safety, efficacy, optimal dosage, and overall impact on patients. Following are some of the important clinical trials that we have summarized.

Carica papaya

In Sri Lanka, twelve patients, who met the eligibility criteria of the clinical trial but declined hospital admission, were administered C. papaya leaf extract (CPLE) in the form of two 5 ml doses at an 8-h interval for adults and two 2.5 ml doses for children under 10 years. All patients showed an increase in platelet counts within 24 h of leaf extract administration, with no hospital admissions required. Some patients with elevated SGPT levels experienced a reduction, and those with low platelet counts and itching hemorrhagic skin rash showed improvement [46].

In a separate case report, C. papaya, was tested in a 45-year-old man bitten by a carrier mosquito. In the investigation, 25 mL of aqueous extract of C. papaya leaves was administered twice daily for five consecutive days. Before the administration, blood samples were analyzed, revealing decreased platelet, white blood cells, and neutrophil count. After the leaves extract administration, a notable increase in platelet (from 55 × 10^3/μL to 168 × 10^3/μL), WBC (from 3.7 × 10^3/μL to 7.7 × 10^3/μL), and neutrophils (from 46.0% to 78.3%) count was observed [6].

In a Malaysian clinical study, C. papaya leaves juice (CPLJ) was investigated for its platelet count-elevating property in patients with dengue fever, and DHF. It was an open-labeled randomized controlled trial involving 228 patients. During the clinical trial, approximately half of the patients received CPLJ for three consecutive days, and the other half served as controls receiving standard management. The results exhibited a significant rise in mean platelet count in the intervention group compared to the control group after 40 and 48 h of admission. Additionally, gene expression studies on ALOX 12 and PTAFR genes revealed high expression in patients on CPLJ, suggesting its potential role in platelet production and aggregation [112].

In another clinical study conducted in Indonesia, investigators used CPLE capsules to assess its platelet improvement potential. In the trial, 80 patients, who met the inclusion criteria, were enrolled for the trial and they were split into two groups (40 patients per group). One cohort served as an intervention cohort, while the other was labeled as a control group. A total of 24 capsules were given to these subjects, divided into twice-daily dosing schedules. The results showed a significantly improved platelet count (p < 0.05), maintained hematocrit stability, and shortened hospital stay of dengue patients in the intervention group [135].

A multicentered clinical trial conducted in Pakistan, Malaysia, Sri Lanka, and other Asian countries investigated CPLE capsules for dengue management. This randomized clinical trial enrolled and split the participants into two groups: a study group receiving CPLE capsules (500 mg) along with routine supportive treatment and a control group receiving only routine supportive treatment. Following the treatment, on the 3rd, 4th, and 5th days, the platelet count in the study group was significantly higher than the control group, in addition to preventing complications of thrombocytopenia in dengue fever [38].

In India, a clinical trial was conducted aimed at assessing the efficacy and safety of CPLE in managing severe thrombocytopenia (≤ 30,000/μL) in adult dengue patients [106]. The extract was utilized in a double-blind, placebo-controlled, randomized, and prospective clinical trial, involving 51 laboratory-confirmed adult dengue patients randomly assigned to either the treatment CPLE group or the placebo group. CPLE-treated patients exhibited a significantly increased platelet count by day 3 compared to the placebo group. Notably, the treatment group also required fewer platelet transfusions and showed a faster recovery to a platelet count of ≥ 50,000/μL. The study suggested that CPLE was safe and well-tolerated, with no significant decrease in mean hospitalization days. Additionally, plasma cytokine profiling indicated potential immunomodulatory effects and a faster clearance kinetics of viral NS1 antigenemia was observed in the CPLE group.

Euphorbia hirta

A clinical trial conducted in Lahore; Pakistan investigated the potential therapeutic effects of the herbal water derived from Euphorbia hirta (Euphorbiaceae) in dengue fever. The trial involved 125 confirmed dengue fever patients. The patients were categorized into two groups based on age. After 24 h of E. hirta herbal water administration, results demonstrated that over 70% of patients exhibited improvement in platelet count, fever, and flu-like symptoms [77] (Table 3).

Table 3 Clinical trials/studies for pharmacologically active medicinal plants DENV infection treatment

Medicinal plants in mix-models (in-vivo and in-vitro)

The following plants have shown anti-dengue effects both in vitro and in vivo studies.

Azadirachta indica

Azadirachta indica, commonly known as Neem, of the family Meliaceae, has been investigated both in vitro and in vivo for anti-dengue activities. Crude aqueous extract of A. indica leaves and pure neem compound (Azadirachtin) were used. The aqueous extract at an MNTD concentration of 1.897 mg/ml completely inhibited the virus in vitro. The in vivo protection studies revealed it inhibited viral replication, as confirmed by the absence of dengue-related clinical symptoms in suckling mice at 120–30 mg/ml [90].

Curcuma longa

In vitro assays of Curcuma longa (Zingiberacea) extract, using Huh7it-1 cells, revealed an IC50 value of 17.91 μg/mL. The selectivity index (SI) calculated from these values was 4.8, indicating a potentially safe and effective antiviral compound [51]. In the in vivo study conducted on ddY mice, oral administration of C. longa extract at a dose of 0.147 mg/mL significantly reduced the viral load after 24 h of DENV-2 infection. Histopathological examination of liver and kidney tissues showed no specific abnormalities and biochemical observations revealed no significant increase in SGPT, SGOT, urea, and creatinine levels. These results suggest that C. longa could serve as a promising antiviral agent against DENV with low cytotoxicity and effective inhibition, highlighting its potential for further exploration and development [51] (Table 2).

Discussion

Plants and Phytocompounds represent a major source of novel therapeutic agents in drug discovery research. Numerous drugs, currently in use, have been fashioned from plant sources. Recently, pharmaceutical companies have started to pour more resources into phytocompound discovery and development owing to technological advances [81]. However, the transition from allopathic to herbal remedies is plagued by various challenges. For instance, naturally sourced drugs have bioavailability issues due to reasons such as poor aqueous solubility and high molecular weight. Moreover, herbal drugs must be validated for their safety and toxicity profile.

With an aim to promote and ensure efficacy, safety and quality of plant-based drugs, the WHO launched a strategy on conventional medicine (2014–23) [56]. In order to optimize the use of and address the drug-delivery challenges associated with phytocompounds, computational screening techniques, such as in silico docking of the therapeutic compound and structure based drug design should be employed [72, 76]. Most of the phytoconstituents revelaed by these approaches have only been evaluated in vitro. To advance the drug development process of phytoconstituents, initially antiviral activity should be tested in a panel of human cell lines that are crucial during natural DENV infection. For example, some of the significant cellular targets during natural dengue virus infection which coule be used for therapeutic evaluation iclude primary human peripheral blood mononuclear cells, hepatic cell lines like Huh7 cells or U2OS cells, and human monocyte-derived macrophages [32].

Moreover, by integrating newly developed organoid cultures, such as 3 dimensional liver organoids, into drug development process could prove to be instrumental to assess the antiviral activity in complex in vitro model [64, 91]. Furthermore, ideally a phytochemical or drug should be active against all serotypes and preferably confirmed for multiple DENV strains within a serotype [69]. Nevertheless, anti-dengue agents or DENV inhibitors which are protective towards two or three serotypes should be promoted to further testing stage. Impotantly, minimal cytotoxicity should be exhibited by these compounds [16]. Novel techniques like cellular thermal shift assays and 3D organoids of the liver may help to gain a more complex and specific insight into the cytotoxicity profile of novel drugs [27, 37].

Lastly, the stability of a phytocompound-based drug is critical for optimal pharmacokinetic profile as limited stability could lead to premature degradation of a therapeutic agent in the gastrointestinal tract or in the liver, significantly impacting the bioavailability [11, 117]. For this purpose, hepatic microsome assay can prove to be more helpful. In this assay, a drug is subjected to subcellular liver fractions with drug-metabolizing enzymes to investigate its metabolic degradation [34, 41, 63]. Ostensibly, it is challenging to identify compounds with the proper characteristics but with thorough research in the virus–host interactions, occurring while viral infection and/or chemical manipulation of identified compounds may accelerate the development of compounds that fit the required criteria [117].

As previously mentioned, lack of proper in vivo testing model also significantly hampers the drug development process. For in vivo testing, The AG129 model is considered the best model to study DENV infection, due to the lack of more representative small animal DENV disease models. AG129 mice are deficient in the interferon I and II receptors, producing high DENV viremia levels and high levels of pro-inflammatory cytokines, leading to thrombocytopenia, vascular leakage, and death [29]. There are two distinct AG129 mouse models, the lethal and the non-lethal mouse model. This is based on the virus strain used and the dose applied [29]. However to study the antiviral efficacy the non-lethal AG129 model is preferred as it allowed to study the time needed to resolve viremia, which was more representative for the clinical situation [117].

It is noteworthy that from 200 viruses identified in humans, only nine can be treated with approved antiviral compounds [29], including human immunodeficiency virus, hepatitis B virus, hepatitis C virus, etc. Numeorous approved regimens are based on combinational therapies, a cocktail of drugs that target different steps of the virus replication cycle [133]. By adding the phytocompounds, especially those that boost platelet count like A. sessillis, C.papaya, more effective anti-dengue regimens can be designed [89]. It is challenging to develop a safe and effective antiviral compound with pan-protective antiviral properties, low toxicity, low chance of viral resistance, and proper stability to ensure absorption and distribution. Many of the plants and phytocompounds, like C.papaya [38, 106] and E. hirta [77] are proving to be effective for dengue treatment in clinical settings by alleviating platelet count. Furthermore, by integrating phytocompounds into the dengue management plans one can hope to not only enhance the therapeutic efficacy but also drive down the overall cost of the treatment [3, 87, 93, 108].

Conclusions

Integrating research on medicinal plants for dengue treatment into a broader agenda is crucial. Efforts are ongoing to establish evidence-based care guidelines for using medicinal plants in dengue infection. It's also important to highlight that the efficacy and safety of medicinal plants in treating dengue virus infection are still areas of ongoing research, and results may vary. The safety and efficacy of these herbal preparations for DSS need more research. A proposed collaborative platform in endemic regions could facilitate studies on fluid therapy and antiviral drug testing of these medicinal plants. Improved surveillance systems in dengue-endemic countries are essential for effective prevention and control. In addition to more research in finding potent cures for dengue infection, vector management strategies should be robust, emphasizing the need to focus on reducing disease incidence.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

DF:

Dengue fever

DENV:

Dengue virus

NK:

Natural killer

IFN:

Interferon

TLRs:

Toll-like receptors

CST:

Castanospermine

DHF:

Dengue hemorrhagic fever

DALYs:

Disability-adjusted life years

DSS:

Dengue shock syndrome

DCs:

Dendritic cells

ISGs:

Interferon-stimulating genes

hCFs:

Human cytotoxic factors

EP:

Envelope protein

MP:

Membrane protein

PBMCs:

Peripheral blood mononuclear cells

RNAi:

RNA interference

EA:

Ethyl acetate

PN:

Phytoniosomes

MNTD:

Maximal non-toxic dose

FFURA:

Foci forming unit reduction assays

MTT:

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide

HPLC–DAD-ESI–MS/MS:

High-performance liquid chromatography-diode array detector-electrospray ionization tandem mass spectrometry

CPLE:

C. papaya Leaf extract

CPLJ:

C. papaya Leaf juice

ADE:

Antibody-Dependent Enhancement

DAAs:

Direct-acting antivirals

HDAs:

Host-directed antivirals

EC:

Endothelial Cell

VE:

Vaccine Efficacy

VCD:

Virologically Confirmed Dengue

References

  1. Abd Wahab NZ, Ibrahim N. Efficacy of catharanthus roseus extract against dengue virus type 2 infection in vitro. Indian J Public Health Res Dev. 2020;11(1):1320–25.

  2. Afandi A, Adib YL, Kee SC, Peng SL, Khai VL, Sanghiran RO, Othman S, Abdul NR, Yusof R, Han CH. Discovery of Dengue Virus Inhibitors. Curr Med Chem. 2020;27(30):4945–5036. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/0929867326666181204155336.

    Article  CAS  Google Scholar 

  3. Abidin SA, Makpol S, Ahmad NS, Nik NA, Muhamad, and Wei Boon Yap. A Review on Unleashing the Potentials of Natural Products in Managing Dengue. Journal of Complementary Medicine Research. 2023;14(2):70–70.

    Article  CAS  Google Scholar 

  4. Acosta EG, Bartenschlager R. The quest for host targets to combat dengue virus infections. Curr Opin Virol. 2016;20:47–54.

    Article  PubMed  Google Scholar 

  5. Afzal HS, Khan W, Nawaz S, Utra AM, Ahmad T. Investigation of the Diuretic Potential of Diosmetin, a Flavonoid from Citrus lemon in Sprague-Dawley Rat Model. Phytopharmacological Communications. 2022;2(02):115–26.

    Article  Google Scholar 

  6. Ahmad N, Fazal H, Ayaz M, Abbasi BH, Mohammad I, Fazal L. Dengue fever treatment with Carica papaya leaves extracts. Asian Pac J Trop Biomed. 2011;1(4):330–3.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Ahmed M, Khan ST. Implications of COVID-19 for Mental Health among Different Sections of the Society. Molecular Medicine Communications. 2022;2(02):157–72.

    Article  Google Scholar 

  8. Ahmed S, Gul S, Gul H, Zia-Ul-Haq M, Ercisli S. Cyclooxygenase-2 inhibition improves antioxidative defense during experimental hypercholesterolemia. Bosn J Basic Med Sci. 2014;14(2):63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Akshatha HS, Gurubasavaraj V, Pujar AK, Sethu MB, Singh M. Dengue structural proteins as antiviral drug targets: Current status in the drug discovery & development. Eur J Med Chem. 2021;221: 113527. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejmech.2021.113527.

    Article  CAS  Google Scholar 

  10. Ali A, Habib ur Rehman, Muhammad Nisar, Shazia Rafique, Sadia Ali, Abrar Hussain, Muhammad Idrees, Sabeen Sabri, Hakeem Zada, and Shah Hussain. Seroepidemiology of dengue fever in Khyber Pakhtunkhawa, Pakistan. Int J Infect Dis. 2013;17(7):e518–23.

    Article  PubMed  Google Scholar 

  11. Ali NA, Dash KK, Pandey VK, Tripathi A, Mukarram SA, Harsányi E, Kovács B. Extraction and Encapsulation of Phytocompounds of Poniol Fruit via Co-Crystallization: Physicochemical Properties and Characterization. Molecules. 2023;28(12):4764.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Aliaga‐Samanez A, Romero D, Murray K, Cobos‐Mayo M, Segura M, Real R, Olivero J.  Climate change is aggravating dengue and yellow fever transmission risk. Ecography. 2024;2024:e06942.

  13. Anasir MI, Ramanathan B, Poh CL. Structure-Based Design of Antivirals against Envelope Glycoprotein of Dengue Virus. Viruses. 2020;12(4):367.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Arollado EC. Platelet augmentation activity of selected Philippine plants. Int J Pharm Phytopharm Res. 2014:3(2):121–3.

  15. Aslam R, Saeed SA, Ahmed S, Connor JD. Lipoproteins inhibit platelet aggregation and arachidonic acid metabolism in experimental hypercholesterolaemia. Clin Exp Pharmacol Physiol. 2008;35:656–62.

  16. Bácskay I, Nemes D, Fenyvesi F, Váradi J, Vasvári G, Fehér P, Vecsernyés M, Ujhelyi Z. Role of cytotoxicity experiments in pharmaceutical development. Cytotoxicity. 2018;8:131–46.

    Google Scholar 

  17. Barboza RS, Valente LMM, Wolff T, Assunção-Miranda I, Neris RLS, Guimarães-Andrade IP, Gomes M. Antiviral activity of Faramea hyacinthina and Faramea truncata leaves on dengue virus type-2 and their major compounds. Chem Biodivers. 2018;15(2): e1700393.

    Article  Google Scholar 

  18. Basavannacharya C, Vasudevan SG. Suramin inhibits helicase activity of NS3 protein of dengue virus in a fluorescence-based high throughput assay format. Biochem Biophys Res Commun. 2014;453(3):539–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrc.2014.09.113.

    Article  CAS  PubMed  Google Scholar 

  19. Bhatt P, Sabeena SP, Varma M, Arunkumar G. Current Understanding of the Pathogenesis of Dengue Virus Infection. Curr Microbiol. 2021;78(1):17–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00284-020-02284-w.

    Article  CAS  PubMed  Google Scholar 

  20. Biswal S, Borja-Tabora C, Vargas LM, Velásquez H, Alera MT, Sierra V, Rodriguez-Arenales EJ, Delia Yu, Pujitha Wickramasinghe V, Moreira ED. Efficacy of a tetravalent dengue vaccine in healthy children aged 4–16 years: a randomised, placebo-controlled, phase 3 trial. The Lancet. 2020;395(10234):1423–33.

    Article  CAS  Google Scholar 

  21. Biswal S, Galvan JFM, Parra MM, Galan-Herrera JF, Rodriguez MBC, Bueno EPR, Brose M, Rauscher M, LeFevre I, Wallace D. Immunogenicity and safety of a tetravalent dengue vaccine in dengue-naïve adolescents in Mexico City. Rev Panam Salud Pública. 2021;45:e6.

  22. Biswal S, Reynales H, Saez-Llorens X, Lopez P, Borja-Tabora C, Kosalaraksa P, Sirivichayakul C, Watanaveeradej V, Rivera L, Espinoza F. Efficacy of a tetravalent dengue vaccine in healthy children and adolescents. N Engl J Med. 2019;381(21):2009–19.

    Article  CAS  PubMed  Google Scholar 

  23. Blackley S, Kou Z, Chen H, Quinn M, Rose RC, Schlesinger JJ, Coppage M, Jin X. Primary human splenic macrophages, but not T or B cells, are the principal target cells for dengue virus infection in vitro. J Virol. 2007;81(24):13325–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Boonnak K, Slike BM, Burgess TH, Mason RM, Shuenn-Jue Wu, Sun P, Porter K, Rudiman IF, Yuwono D, Puthavathana P, Marovich MA. Role of Dendritic Cells in Antibody-Dependent Enhancement of Dengue Virus Infection. J Virol. 2008;82(8):3939–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/jvi.02484-07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Botta L, Rivara M, Zuliani V, Radi M. Drug repurposing approaches to fight Dengue virus infection and related diseases. Front Biosci (Landmark Ed). 2018;23(6):997–1019.

    CAS  PubMed  Google Scholar 

  26. Bourjot M, Leyssen P, Eydoux C, Guillemot JC, Canard B, Rasoanaivo P, Guéritte F, Litaudon M. Flacourtosides A-F, phenolic glycosides isolated from Flacourtia ramontchi. J Nat Prod. 2012;75(4):752–8.

    Article  CAS  PubMed  Google Scholar 

  27. Brooks A, Liang X, Zhang Y, Zhao C-X, Roberts MS, Wang H, Zhang L, Crawford DHG. Liver organoid as a 3D in vitro model for drug validation and toxicity assessment. Pharmacol Res. 2021;169: 105608.

    Article  CAS  PubMed  Google Scholar 

  28. Bundeesomchok K, Filly A, Rakotomanomana N, Panichayupakaranant P, Chemat F. Extraction of α-mangostin from Garcinia mangostana L. using alternative solvents: Computational predictive and experimental studies. LWT Food Sci Technol. 2016;65:297–303. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lwt.2015.08.036.

    Article  CAS  Google Scholar 

  29. Chan KW, Ki SW, Kavishna R, Alonso S, Vasudevan SG. Animal models for studying dengue pathogenesis and therapy. Antiviral Res. 2015;123:5–14.

    Article  CAS  PubMed  Google Scholar 

  30. Chao CH, Wei-Chueh Wu, Lai YC, Tsai PJ, Perng GC, Lin YS, Yeh TM. Dengue virus nonstructural protein 1 activates platelets via Toll-like receptor 4, leading to thrombocytopenia and hemorrhage. PLoS Pathog. 2019;15(4): e1007625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chappell JD, Dermody TS. Biology of viruses and viral diseases. In: Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 2015. p. 1681.

    Chapter  Google Scholar 

  32. Chen YL, Ghafar NA, Karuna R, Yilong Fu, Lim SP, Schul W, Feng Gu, Herve M, Yokohama F, Wang G, Cerny D, Fink K, Blasco F, Shi P-Y. Activation of Peripheral Blood Mononuclear Cells by Dengue Virus Infection Depotentiates Balapiravir. J Virol. 2014;88(3):1740–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/jvi.02841-13.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Chiow KH, Phoon MC, Putti T, Tan BKH, Chow VT. Evaluation of antiviral activities of Houttuynia cordata Thunb. extract, quercetin, quercetrin and cinanserin on murine coronavirus and dengue virus infection. Asian Pac J Trop Med. 2016;9(1):1–7.

    Article  CAS  PubMed  Google Scholar 

  34. Di Li, Kerns EH, Ma XJ, Huang Y, Carter GT. Applications of high throughput microsomal stability assay in drug discovery. Comb Chem High Throughput Screening. 2008;11(6):469–76.

    Article  CAS  Google Scholar 

  35. Durbin AP, Vargas MJ, Wanionek K, Hammond SN, Gordon A, Rocha C, Balmaseda A, Harris E. Phenotyping of peripheral blood mononuclear cells during acute dengue illness demonstrates infection and increased activation of monocytes in severe cases compared to classic dengue fever. Virology. 2008;376(2):429–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.virol.2008.03.028.

    Article  CAS  PubMed  Google Scholar 

  36. Espina LM, Valero NJ, Hernandez JANETHM, Mosquera JA. Increased apoptosis and expression of tumor necrosis factor-alpha caused by infection of cultured human monocytes with dengue virus. Am J Trop Med Hyg. 2003;68(1):48–53.

    Article  PubMed  Google Scholar 

  37. Foglietta F, Canaparo R, Muccioli G, Terreno E, Serpe L. Methodological aspects and pharmacological applications of three-dimensional cancer cell cultures and organoids. Life Sci. 2020;254: 117784.

    Article  CAS  PubMed  Google Scholar 

  38. Gadhwal AK, Ankit BS, Chahar C, Pankaj Tantia P, Sirohi, and RP Agrawal. Effect of Carica papaya Leaf Extract Capsule on Platelet Count in Patients of Dengue Fever with Thrombocytopenia. J Assoc Physicians India. 2016;64(6):22–6.

    PubMed  Google Scholar 

  39. Ghany MG, Watkins PB. Moving the Needle to Reduce Acetaminophen (Paracetamol) Hepatotoxicity. JAMA. 2023;329(9):713–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jama.2023.1002.

    Article  CAS  PubMed  Google Scholar 

  40. Glasner DR, Henry Puerta-Guardo P, Beatty R, Harris E. The good, the bad, and the shocking: the multiple roles of dengue virus nonstructural protein 1 in protection and pathogenesis. Annual review of virology. 2018;5(1):227–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Godoi AB, de Jesus N, Antunes KF, Cunha AF, Martins MA, Huestis, and Jose Luiz Costa. Metabolic Stability and Metabolite Identification of N-Ethyl Pentedrone Using Rat, Mouse and Human Liver Microsomes. Pharmaceutics. 2024;16(2):257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gul S, Ahmed S, Gul H, Shad KF, Zia-Ul-Haq M, Badiu D. The antioxidant potential of Brassica rapa L. on glutathione peroxidase, superoxide dismutase enzymes and total antioxidant status. Revista Romana de Medicina de Laborator. 2013;21(2):161–9.

    Article  Google Scholar 

  43. Halstead SB. Delivering safe dengue vaccines. Lancet Glob Health. 2024;12(8):e1229–30.

    Article  CAS  PubMed  Google Scholar 

  44. Harapan H, Alice Michie R, Sasmono T, Imrie A. Dengue: a minireview. Viruses. 2020;12(8):829.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Harish V, Colón-González FJ, Moreira FRR, Gibb R, Kraemer MUG, Davis M, Reiner RC, Pigott DM, Alex Perkins T, Weiss DJ, Bogoch II, Vazquez-Prokopec G, Saide PM, Barbosa GL, Sabino EC, Khan K, Faria NR, Hay SI, Correa-Morales F, Chiaravalloti-Neto F, Brady OJ. Human movement and environmental barriers shape the emergence of dengue. Nat Commun. 2024;15(1):4205. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-024-48465-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hettige S. Salutary effects of Carica papaya leaf extract in dengue fever patients–a pilot study. Sri Lankan Family Physician. 2008;29(1):17–9.

    Google Scholar 

  47. Huerre MR, Lan NT, Marianneau P, Hue NB, Khun H, Hung NT, Khen NT, Drouet MT, Huong VTQ, Ha DQ, Buisson Y, Deubel V. Liver histopathology and biological correlates in five cases of fatal dengue fever in Vietnamese children. Virchows Arch. 2001;438(2):107–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s004280000329.

    Article  CAS  PubMed  Google Scholar 

  48. Huerta-Zepeda A, Cabello-Gutiérrez C, Cime-Castillo J, Monroy-Martínez V, Manjarrez-Zavala ME, Gutiérrez-Rodríguez M, Izaguirre R, Ruiz-Ordaz BH. Crosstalk between coagulation and inflammation during Dengue virus infection. Thromb Haemost. 2008;99(11):936–43.

    Article  CAS  PubMed  Google Scholar 

  49. Hussain J, Khan FU, Gilani SA, Abbas G, Ahmed S, Khan AU, Ullah W, Choudhary MI.  Antiglycation, antiplatelets aggregation, cytotoxic and phytotoxic activities of Nepeta suavis. Lat Am J Pharm. 2010;29:573–8.

  50. Hussain Z, Rani S, Ma F, Li W, Shen W, Gao T, Wang J, Pei R. Dengue determinants: Necessities and challenges for universal dengue vaccine development. Rev Med Virol. 2023;33(2): e2425. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/rmv.2425.

    Article  CAS  PubMed  Google Scholar 

  51. Ichsyani M, Ridhanya A, Risanti M, Desti H, Ceria R, Putri DH, Sudiro TM, Dewi BE. Antiviral effects of Curcuma longa L. against dengue virus in vitro and in vivo. In: Paper read at IOP Conference Series: Earth and Environmental Science. 2017.

    Google Scholar 

  52. IFRC. Ministry of National Health Services Regulation and Coordination (NHSRC) of Pakistan. 2019.

    Google Scholar 

  53. Imran I, Hussain L, Ahmed S, Rasool N, Rasool S, Abbas G, Ali MY. Antiplatelet activity of methanolic extract of Acacia leucophloea bark. J Med Plants Res. 2012;6(25):4185–8.

    Google Scholar 

  54. Iqtadar S, Akram J, Khan A. The Urgent Need for Dengue Vaccination: Combating an Escalating Public Health Crisis in Pakistan. Vaccines. 2024;12(8):913.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Jain M, Ganju L, Katiyal A, Padwad Y, Mishra KP, Chanda S, Karan D, Yogendra KMS, Sawhney RC. Effect of Hippophae rhamnoides leaf extract against Dengue virus infection in human blood-derived macrophages. Phytomedicine. 2008;15(10):793–9.

    Article  CAS  PubMed  Google Scholar 

  56. Jha, Shikha, Klaudi K. Vaiphei, and Amit Alexander. "Chapter 1 - Plant-based therapeutics: current status and future perspectives." In Phytopharmaceuticals and Herbal Drugs, edited by Manju Rawat Singh and Deependra Singh, 3–11. Cambridge: Academic Press; 2023.

  57. Jiang WL, Luo XL, Kuang SJ. Effects of Alternanthera philoxeroides Griseb against dengue virus in vitro. Di Yi Jun Yi Da Xue Xue Bao. 2005;25(4):454–6.

    PubMed  Google Scholar 

  58. Kamath V, Aishwarya AB. Dengue Vaccines: Current Status and Future Perspectives. APIK Journal of Internal Medicine. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/ajim.ajim_108_23.10.4103/ajim.ajim_108_23.

    Article  Google Scholar 

  59. Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host-directed therapies for bacterial and viral infections. Nat Rev Drug Discovery. 2018;17(1):35–56.

    Article  CAS  PubMed  Google Scholar 

  60. Khan MB, Yang Z-S, Lin C-Y, Hsu M-C, Urbina AN, Assavalapsakul W, Wang W-H, Chen Y-H, Wang S-F. Dengue overview: An updated systemic review. J Infect Public Health. 2023;16(10):1625–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jiph.2023.08.001.

    Article  PubMed  Google Scholar 

  61. Khan S, Najeeb ur Rehman, Amanullah Lail, and Seema Aftab. Differential Viral Load Indicates HDV Influences HBV Replication in HBV/HDV Coinfected Patients. Molecular Medicine Communications. 2022;2(02):149–56.

    Article  Google Scholar 

  62. Klawikkan N, Nukoolkarn V, Jirakanjanakit N, Yoksan S, Wiwat C. Effect of Thai medicinal plant extracts against Dengue virus in vitro. 2010.

    Google Scholar 

  63. Knights KM, Stresser DM, Miners JO, Crespi CL. In Vitro Drug Metabolism Using Liver Microsomes. Curr Protoc Pharmacol. 2016;74(1):7.8.1–7.8.24. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cpph.9.

    Article  PubMed  Google Scholar 

  64. Kronemberger GS, Carneiro FA, Rezende DF, Baptista LS. Spheroids and organoids as humanized 3D scaffold-free engineered tissues for SARS-CoV-2 viral infection and drug screening. Artif Organs. 2021;45(6):548–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/aor.13880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kularatne SA, Dalugama C. Dengue infection: Global importance, immunopathology and management. Clin Med. 2022;22(1):9.

    Article  Google Scholar 

  66. Leardkamolkarn V, Sirigulpanit W, Phurimsak C, Kumkate S, Himakoun L, Sripanidkulchai B. The inhibitory actions of Houttuynia cordata aqueous extract on dengue virus and dengue-infected cells. J Food Biochem. 2012;36(1):86–92.

    Article  CAS  Google Scholar 

  67. Lee SH, Tang YQ, Rathkrishnan A, Wang SM, Ong KC, Manikam R, Payne BJ, Jaganath IB, Sekaran SD. Effects of cocktail of four local Malaysian medicinal plants (Phyllanthus spp.) against dengue virus 2. BMC Complement Altern Med. 2013;13:1–13.

    Article  Google Scholar 

  68. Li Q, Kang C. Dengue virus NS4B protein as a target for developing antivirals. Front Cell Infect Microbiol. 2022;12. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcimb.2022.959727.

  69. Lim SP. Dengue drug discovery: Progress, challenges and outlook. Antiviral Res. 2019;163:156–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.antiviral.2018.12.016.

    Article  CAS  PubMed  Google Scholar 

  70. Lim SP, Wang Q-Y, Noble CG, Chen Y-L, Dong H, Zou B, Yokokawa F, Nilar S, Smith P, Beer D. Ten years of dengue drug discovery: progress and prospects. Antiviral Res. 2013;100(2):500–19.

    Article  CAS  PubMed  Google Scholar 

  71. Limon-Flores AY, Perez-Tapia M, Estrada-Garcia I, Vaughan G, Escobar-Gutierrez A, Calderon-Amador J, Herrera-Rodriguez SE, Brizuela-Garcia A, Heras-Chavarria M, Flores-Langarica A, Cedillo-Barron L, Flores-Romo L. Dengue virus inoculation to human skin explants: an effective approach to assess in situ the early infection and the effects on cutaneous dendritic cells. Int J Exp Pathol. 2005;86(5):323–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.0959-9673.2005.00445.x.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Low JG, Gatsinga R, Vasudevan SG, Sampath A. Dengue antiviral development: a continuing journey. Dengue and Zika: Control and Antiviral Treatment Strategies. 2018;1062:319–332.

  73. Luplerdlop N, Missé D, Bray D, Deleuze V, Gonzalez JP, Leardkamolkarn V, Yssel H, Veas F. Dengue-virus-infected dendritic cells trigger vascular leakage through metalloproteinase overproduction. EMBO Rep. 2006;7(11):1176–81.

    Article  CAS  PubMed Central  Google Scholar 

  74. Martina BE, Koraka P, Osterhaus AD. Dengue virus pathogenesis: an integrated view. Clin Microbiol Rev. 2009;22(4):564–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/cmr.00035-09.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Mehmood Y, Arshad M. Dengue in the urban slums of Pakistan: health costs, adaptation practices, and the role of dengue-diagnosis and surveillance in controlling the epidemic. GeoJournal. 2024;89(2):75. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10708-024-11073-y.

    Article  Google Scholar 

  76. Miller JL, Lachica R, Sayce AC, Williams JP, Bapat M, Raymond Dwek P, Beatty R, Harris E, Zitzmann N. Liposome-mediated delivery of iminosugars enhances efficacy against dengue virus in vivo. Antimicrob Agents Chemother. 2012;56(12):6379–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mir M, Khurshid R, Aftab R. Management of thrombocytopenia and flu-like symptoms in dengue patients with herbal water of Euphorbia hirta. J Ayub Med Coll Abbottabad. 2012;24(3–4):6–9.

    PubMed  Google Scholar 

  78. Mulakoli F, Gachara G, Ndombi E, Khamadi S. Dengue Virus and Blood Safety: A Mini-Review of Research Publications. East Africa Science. 2023;5(1):1–7.

    Article  Google Scholar 

  79. Muliawan SY, Kit LS, Devi S, Hashim O, Yusof R. Inhibitory potential of Quercus lusitanica extract on dengue virus type 2 replication. The Southeast Asian Journal of Tropical Medicine and Public Health. 2006;37:132–5.

    PubMed  Google Scholar 

  80. Muller DA, Young PR. The flavivirus NS1 protein: molecular and structural biology, immunology, role in pathogenesis and application as a diagnostic biomarker. Antiviral Res. 2013;98(2):192–208.

    Article  CAS  PubMed  Google Scholar 

  81. Najmi A, Javed SA, Bratty MA, Alhazmi HA. Modern Approaches in the Discovery and Development of Plant-Based Natural Products and Their Analogues as Potential Therapeutic Agents. Molecules. 2022;27(2):349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Nakao S, Lai C-J, Young NS. Dengue virus, a flavivirus, propagates in human bone marrow progenitors and hematopoietic cell lines. Blood. 1989;74(4):1235–40.

    Article  CAS  PubMed  Google Scholar 

  83. Nasar S, Rashid N, Iftikhar S. Dengue proteins with their role in pathogenesis, and strategies for developing an effective anti-dengue treatment: A review. J Med Virol. 2020;92(8):941–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jmv.25646.

    Article  CAS  PubMed  Google Scholar 

  84. Nascimento AC, Valente LMM, Gomes M, Barboza RS, Wolff T, Neris RLS, Figueiredo CM, Assunção-Miranda I. Antiviral activity of Faramea bahiensis leaves on dengue virus type-2 and characterization of a new antiviral flavanone glycoside. Phytochem Lett. 2017;19:220–5.

    Article  CAS  Google Scholar 

  85. Franco EJ, Drusano GL, Hanrahan KC, Warfield KL, Brown AN. Combination Therapy with UV-4B and Molnupiravir Enhances SARS-CoV-2 Suppression. Viruses. 2023;15(5):1175. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/v15051175. https://clinicaltrials.gov/study/NCT02061358.

  86. Nguyen PT, Viet GL, Nguyen T, Đinh OT, Duong CQ, Nguyen LH, Truong TN. In search of suitable protein targets for anti-malarial and anti-dengue drug discovery. J Mol Struct. 2022;1256: 132520. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molstruc.2022.132520.

    Article  CAS  Google Scholar 

  87. Ogbole OO, Akinleye TE, Segun PA, Faleye TC, Adeniji AJ. In vitro antiviral activity of twenty-seven medicinal plant extracts from Southwest Nigeria against three serotypes of echoviruses. Virology Journal. 2018;15(1):110. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12985-018-1022-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Osorio JE, Wallace D, Stinchcomb DT. A recombinant, chimeric tetravalent dengue vaccine candidate based on a dengue virus serotype 2 backbone. Expert Rev Vaccines. 2016;15(4):497–508.

    Article  CAS  PubMed  Google Scholar 

  89. Panday H, Jha AK, Jha NK, Jha SK. Role of dietary supplements: post dengue complications. J Sci Tech Res. 2021;11(2):1–8.

  90. Parida MM, Upadhyay C, Pandya G, Jana AM. Inhibitory potential of neem (Azadirachta indica Juss) leaves on dengue virus type-2 replication. J Ethnopharmacol. 2002;79(2):273–8.

    Article  CAS  PubMed  Google Scholar 

  91. Park J, Wetzel I, Dréau D, Cho H. 3D miniaturization of human organs for drug discovery. Adv Healthcare Mater. 2018;7(2):1700551.

    Article  Google Scholar 

  92. Parveen S, Riaz Z, Saeed S, Ishaque U, Sultana M, Faiz Z, Shafqat Z, Shabbir S, Ashraf S, Marium A. Dengue hemorrhagic fever: a growing global menace. J Water Health. 2023;21(11):1632–50. https://doiorg.publicaciones.saludcastillayleon.es/10.2166/wh.2023.114.

    Article  PubMed  Google Scholar 

  93. Paul A, Chakraborty N, Sarkar A, Acharya K, Ranjan A, Chauhan A, Srivastava S, Singh AK, Rai AK, Mubeen I, Prasad R. Ethnopharmacological Potential of Phytochemicals and Phytogenic Products against Human RNA Viral Diseases as Preventive Therapeutics. Biomed Res Int. 2023;2023(1):1977602. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2023/1977602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Pintado Silva J, Fernandez-Sesma A. Challenges on the development of a dengue vaccine: a comprehensive review of the state of the art. J Gen Virol. 2023;104(3). https://doiorg.publicaciones.saludcastillayleon.es/10.1099/jgv.0.001831.

  95. Pong LY, Yew PN, Lee WL, Lim YY, Sharifah SH. Anti-dengue virus serotype 2 activity of tannins from porcupine dates. Chinese Medicine. 2020;15(1):49. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13020-020-00329-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Raafat N, Blacksell SD, Maude RJ. A review of dengue diagnostics and implications for surveillance and control. Trans R Soc Trop Med Hyg. 2019;113(11):653–60.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Ramalingam S, Karupannan S, Padmanaban P, Vijayan S, Sheriff K, Palani G, Kaveri K, Krishnasamy. Anti-dengue activity of Andrographis paniculata extracts and quantification of dengue viral inhibition by SYBR green reverse transcription polymerase chain reaction. Ayu. 2018;39(2):87.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Ramu ST, Ariyaratne D, Jeewandara C, Jayathilaka D, Gomes L, Wijewickrama A, Ogg G, Malavige GN. Identifying an Immunodominant Epitope of Dengue Non-Structural Protein 1 (NS1) in Acute Secondary Dengue Infections for Vaccine Development or Development of Treatment Modalities. Paper read at 2020 From Innovation to Impact (FITI). 2020. p. 15.

    Google Scholar 

  99. Rehman NU, Begum N, Ali L, Al-Harrasi A, Abbas G, Sagheer Ahmad ABDULLATIF, Khan ZK, Shinwari, and Javid Hussain. Lipid peroxidation, antiglycation, cytotoxic, phytotoxic, antioxidant, antiplatelet and antimicrobial activities of Ajuga bracteosa against various pathogens. Pak J Bot. 2015;47(3):1195–7.

    CAS  Google Scholar 

  100. Reis SR, IN, Ligia MM Valente, André L Sampaio, Antonio C Siani, Mariana Gandini, Elzinandes L Azeredo, Luiz A D’Avila, José L Mazzei, M Henriques Maria das Graças, and Claire F Kubelka. Immunomodulating and antiviral activities of Uncaria tomentosa on human monocytes infected with Dengue Virus-2. Int Immunopharmacol. 2008;8(3):468–76.

    Article  CAS  PubMed  Google Scholar 

  101. Rosmalena R, Elya B, Dewi BE, Fithriyah F, Desti H, Angelina M, Hanafi M, Lotulung PD, Prasasty VD, Seto D. The antiviral effect of indonesian medicinal plant extracts against dengue virus in vitro and in silico. Pathogens. 2019;8(2):85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Saleem N, Shahid T. Search for Natural Remedies for the Treatment of Salmonella Typhi Infections. Microbiological & Immunological Communications. 2023;2(02):121–34.

    Article  Google Scholar 

  103. Samsa MM, Mondotte JA, Caramelo JJ, Gamarnik AV. Uncoupling <i>cis</i>-Acting RNA Elements from Coding Sequences Revealed a Requirement of the N-Terminal Region of Dengue Virus Capsid Protein in Virus Particle Formation. J Virol. 2012;86(2):1046–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/jvi.05431-11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Sarker R, Roknuzzaman ASM, Aminul Haque Md, Rabiul Islam Md, Kabir ER. Upsurge of dengue outbreaks in several WHO regions: Public awareness, vector control activities, and international collaborations are key to prevent spread. Health Science Reports. 2024;7(4): e2034. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hsr2.2034.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Sathasivam K, Ramanathan S, Mansor SM, Haris MRMH, Wernsdorfer WH. Thrombocyte counts in mice after the administration of papaya leaf suspension. Wiener klinische Wochenschrift. 2009;121:19–22.

  106. Sathyapalan DT, Padmanabhan A, Moni M, Binny P-Prabhu, Preetha Prasanna, Sabarish Balachandran, Sreekrishnan P Trikkur, Soumya Jose, Fabia Edathadathil, and Jagan O Anilkumar. Efficacy & safety of Carica papaya leaf extract (CPLE) in severe thrombocytopenia (≤ 30,000/μl) in adult dengue–Results of a pilot study. PLoS ONE. 2020;15(2): e0228699.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Shanmugam A, Ramakrishnan C, Velmurugan D, Gromiha MM. Identification of Potential Inhibitors for Targets Involved in Dengue Fever. Curr Top Med Chem. 2020;20(19):1742–60. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1568026620666200618123026.

    Article  CAS  PubMed  Google Scholar 

  108. Sharma AN, Dewangan HK, Upadhyay PK. Comprehensive Review on Herbal Medicine: Emphasis on Current Therapy and Role of Phytoconstituents for Cancer Treatment. Chem Biodivers. 2024;21(3): e202301468. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cbdv.202301468.

    Article  CAS  PubMed  Google Scholar 

  109. Silva ARA, Morais SM, Marques MMM, Lima DM, Santos SCC, Almeida RR, Vieira IGP, Guedes MIF. Antiviral activities of extracts and phenolic components of two Spondias species against dengue virus. Journal of Venomous Animals and Toxins Including Tropical Diseases. 2011;17:406–13.

    CAS  Google Scholar 

  110. Sirivichayakul, Chukiat, Shibadas Biswal, Xavier Saez-Llorens, Eduardo López-Medina, Charissa Borja-Tabora, Lulu Bravo, Pope Kosalaraksa, Maria Theresa Alera, Humberto Reynales, Luis Rivera, Veerachai Watanaveeradej, Delia Yu, Felix Espinoza, Reynaldo Dietze, LakKumar Fernando, V Pujitha Wickramasinghe, Edson Duarte Moreira, Jr, Asvini D Fernando, Dulanie Gunasekera, Kleber Luz, Rivaldo Venâncio da Cunha, Ana Lucia Oliveira, Martina Rauscher, Huihao Fan, Astrid Borkowski, Ian Escudero, Suely Tuboi, Eric Lloyd, Vianney Tricou, Nicolas Folschweiller, Inge LeFevre, Luis Martinez Vargas, Derek Wallace, and for the TIDES Study Group. Efficacy and Safety of a Tetravalent Dengue Vaccine (TAK-003) in Children With Prior Japanese Encephalitis or Yellow Fever Vaccination. J Infect Dis. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/infdis/jiae222.

    Article  Google Scholar 

  111. Sreekanth GP, Chuncharunee A, Yenchitsomanus P-T, Limjindaporn T. Crocetin Improves Dengue Virus-Induced Liver Injury. Viruses. 2020;12(8):825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Subenthiran S, Choon TC, Cheong KC, Thayan R, Teck MB, Muniandy PK, Afzan A, Abdullah NR, Ismail Z. Carica papaya leaves juice significantly accelerates the rate of increase in platelet count among patients with dengue fever and dengue haemorrhagic fever. Evid Based Complement Alternat Med. 2013;2013:616737.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Tabassum S, Naeem A, Nazir A, Naeem F, Gill S, Tabassum S. Year-round dengue fever in Pakistan, highlighting the surge amidst ongoing flood havoc and the COVID-19 pandemic: a comprehensive review. Annals of Medicine and Surgery. 2023;85(4):908–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/ms9.0000000000000418.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Tayone WC, Tayone JC, Hashimoto M. Isolation and structure elucidation of potential Anti-Dengue metabolites from Tawa-Tawa (Euphorbia hirta Linn.).  Walailak J Sci Technol (WJST). 2014.;11(10):825–832.

  115. Thomas SJ. Is new dengue vaccine efficacy data a relief or cause for concern?. NPJ Vacc. 2023;8(1):55. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41541-023-00658-2.

  116. Tricou V, Delia Yu, Reynales H, Biswal S, Saez-Llorens X, Sirivichayakul C, Lopez P, Borja-Tabora C, Bravo L, Kosalaraksa P. Long-term efficacy and safety of a tetravalent dengue vaccine (TAK-003): 4· 5-year results from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Glob Health. 2024;12(2):e257–70.

    Article  CAS  PubMed  Google Scholar 

  117. Troost B, Smit JM. Recent advances in antiviral drug development towards dengue virus. Curr Opin Virol. 2020;43:9–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.coviro.2020.07.009.

    Article  CAS  PubMed  Google Scholar 

  118. Trujillo-Correa AI, Quintero-Gil DC, Diaz-Castillo F, Quiñones W, Robledo SM, Martinez-Gutierrez M. In vitro and in silico anti-dengue activity of compounds obtained from Psidium guajava through bioprospecting. BMC Complement Altern Med. 2019;19:1–16.

    Article  CAS  Google Scholar 

  119. Vidanapathirana M, Atukorala I. Dengue hemorrhagic fever with bleeding and fluid overload in a patient with active lupus nephritis: a case report of diagnostic and therapeutic challenges. BMC Infect Dis. 2023;23(1):433. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12879-023-08415-5.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Wahab NZA, Ibrahim N, Kamarudin MKA, Lananan F, Juahir H, Ghazali A. In vitro antiviral activity of Orthosiphon stamineus extract against dengue virus type 2. Journal of Fundamental and Applied Sciences. 2018;10(1S):541–51.

    CAS  Google Scholar 

  121. Wahab NZA, Ibrahim N, Kamarudin MKA, Lananan F, Juahir H, Ghazali A, Ireana AF, Yusra. Cytotoxicity and antiviral activity of Annona muricata aqueous leaves extract against dengue virus type 2. Journal of Fundamental and Applied Sciences. 2018;10(1S):580–9.

    CAS  Google Scholar 

  122. WHO. Dengue fever – Pakistan. 2021.

    Google Scholar 

  123. WHO. Dengue - Global situation. 2024.

    Google Scholar 

  124. WHO, mondiale de la Santé. WHO position paper on dengue vaccines–May 2024= Note de synthèse: position de l’OMS sur les vaccins contre la dengue–mai 2024. Wkly Epidemiol Rec. 2024;99:203–224.

  125. Wilder-Smith A, Ooi E-E, Horstick O, Wills B. Dengue. The Lancet. 2019;393(10169):350–63.

    Article  Google Scholar 

  126. Wilson DK, Shyamala G, Paulpandi M, Narayanasamy A, Siram K, Karuppaiah A, Sankar V. Development and characterization of phytoniosome nano vesicle loaded with aqueous leaf extracts of Justicia adhatoda and Psidium guajoava against dengue virus (DEN-2). J Cluster Sci. 2021;32:297–304.

    Article  CAS  Google Scholar 

  127. Wolff T, Berrueta LA, Valente LMM, Barboza RS, Neris RLS, Guimarães-Andrade IP, Assunção-Miranda I, Nascimento AC, Gomes M, Gallo B. Comprehensive characterisation of polyphenols in leaves and stems of three anti-dengue virus type-2 active Brazilian Faramea species (Rubiaceae) by HPLC-DAD-ESI-MS/MS. Phytochem Anal. 2019;30(1):62–72.

    Article  CAS  PubMed  Google Scholar 

  128. Wong JM, Adams LE, Durbin AP, Muñoz-Jordán JL, Poehling KA, Sánchez-González LM, Volkman HR, Paz-Bailey G. Dengue: a growing problem with new interventions. Pediatrics. 2022;149(6): e2021055522.

    Article  PubMed  Google Scholar 

  129. Wu S-J, Grouard-Vogel G, Sun W, Mascola JR, Brachtel E, Putvatana R, Louder MK, Filgueira L, Marovich MA, Wong HK, Blauvelt A, Murphy GS, Robb ML, Innes BL, Birx DL, Hayes CG, Frankel SS. Human skin Langerhans cells are targets of dengue virus infection. Nat Med. 2000;6(7):816–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/77553.

    Article  CAS  PubMed  Google Scholar 

  130. Xie X, Wang QY, Hao Ying Xu, Qing M, Kramer L, Yuan Z, Shi PY. Inhibition of Dengue Virus by Targeting Viral NS4B Protein. J Virol. 2011;85(21):11183–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/jvi.05468-11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Yadav M, Malakar A, Mukherjee P, Mukherjee S, Mahajan T, George N. Medicinal Plants and Herbs in the Treatment of Dengue Virus. In Promising Antiviral Herbal and Medicinal Plants. Boca Raton: CRC Press; 2024. p. 101–120.

  132. Yang X, Quam MBM, Zhang T, Sang S. Global burden for dengue and the evolving pattern in the past 30 years. J Travel Med. 2021;28(8):taab146.

    Article  PubMed  Google Scholar 

  133. Yeo KL, Chen YL, Hao Ying Xu, Dong H, Wang QY, Yokokawa F, Shi PY. Synergistic suppression of dengue virus replication using a combination of nucleoside analogs and nucleoside synthesis inhibitors. Antimicrob Agents Chemother. 2015;59(4):2086–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Yongpitakwattana P, Morchang A, Panya A, Sawasdee N, Yenchitsomanus P-T. Alpha-mangostin inhibits dengue virus production and pro-inflammatory cytokine/chemokine expression in dendritic cells. Adv Virol. 2021;166(6):1623–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00705-021-05017-x.

    Article  CAS  Google Scholar 

  135. Yunita F, Hanani E, Kristianto J. The effect of Carica papaya L. leaves extract capsules on platelets count and hematocrit level in dengue fever patient. Int J Med Aromat Plants. 2012;2(4):573–8.

    Google Scholar 

  136. Zhang Y, Corver J, Chipman PR, Zhang W, Pletnev SV,  Sedlak D, Baker TS, Strauss JH, Kuhn RJ, Rossmann MG. Structures of immature flavivirus particles. EMBO J. 2003;2003:2604–13.

  137. Zohra T, Din M, Ikram A, Bashir A, Jahangir H, Baloch IS, Irshad S, Waris A, Salman M, Iqtadar S, Ayaz M. Demographic and clinical features of dengue fever infection in Pakistan: a cross-sectional epidemiological study. 2024;10 (1):11. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40794-024-00221-4.

Download references

Funding

No funding was sought for this review manuscript.

Author information

Authors and Affiliations

Authors

Contributions

SA and FN conceptualized the study, BR, NS, AA, SK did the literature search and collected data from the studies, performed the critical analysis of the studies and wrote the initial manuscript, SA and FN finalized the manuscript, all authors agreeD with the final manuscript.

Corresponding authors

Correspondence to Faiza Naseer or Sagheer Ahmad.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rehman, B., Ahmed, A., Khan, S. et al. Exploring plant-based dengue therapeutics: from laboratory to clinic. Trop Dis Travel Med Vaccines 10, 23 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40794-024-00232-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40794-024-00232-1

Keywords