Next Article in Journal
Structural Analysis of Inhibitor Binding to Enterovirus-D68 3C Protease
Previous Article in Journal
Genetic and Molecular Characterization of Avian Influenza A(H9N2) Viruses from Live Bird Markets (LBM) in Senegal
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Review of the Development and Therapeutic Use of Antivirals in Flavivirus Infection

by
Aarti Tripathi
1,2,†,
Shailendra Chauhan
1,2,*,† and
Renu Khasa
3,*
1
Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
2
Galveston National Laboratory, Galveston, TX 77555, USA
3
Department of Microbiology and Immunology, Miller School of Medicine, University of Miami/UHealth, Miami, FL 33136, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2025, 17(1), 74; https://doi.org/10.3390/v17010074
Submission received: 9 November 2024 / Revised: 29 December 2024 / Accepted: 30 December 2024 / Published: 8 January 2025
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)

Abstract

:
Flaviviruses are a diverse group of viruses primarily transmitted through hematophagous insects like mosquitoes and ticks. Significant expansion in the geographic range, prevalence, and vectors of flavivirus over the last 50 years has led to a dramatic increase in infections that can manifest as hemorrhagic fever or encephalitis, leading to prolonged morbidity and mortality. Millions of infections every year pose a serious threat to worldwide public health, encouraging scientists to develop a better understanding of the pathophysiology and immune evasion mechanisms of these viruses for vaccine development and antiviral therapy. Extensive research has been conducted in developing effective antivirals for flavivirus. Various approaches have been extensively utilized in clinical trials for antiviral development, targeting virus entry, replication, polyprotein synthesis and processing, and egress pathways exploiting virus as well as host proteins. However, to date, no licensed antiviral drug exists to treat the diseases caused by these viruses. Understanding the mechanisms of host–pathogen interaction, host immunity, viral immune evasion, and disease pathogenesis is highly warranted to foster the development of antivirals. This review provides an extensively detailed summary of the most recent advances in the development of antiviral drugs to combat diseases.

1. Introduction

Survival of the fittest is a law of evolution that applies to all entities, including viruses. Virus infection does not always translate to disease. The disease manifestation depends on persistent viral multiplication and dysregulated host immune response. The development of antiviral compounds against flaviviruses is a critical area of research due to the significant global health burden posed by these viruses, which include dengue virus (DENV), Zika virus (ZIKV), West Nile virus (WNV), Japanese encephalitis virus (JEV), and yellow fever virus (YFV). The flavivirus family has RNA as genetic material amplified by RNA-dependent RNA polymerase (RdRp). Owing to the absence of proof-reading ability, the viral progeny are prone to accumulation of mutations, producing a defective, nonfunctional virus or mutated variants with survival fitness, allowing the virus to hijack the host cellular machinery, thus evading the host immune response. This implies that the viral structural and non-structural proteins play a vital role in virus replication, assembly, and egress that can be targeted to develop antiviral therapeutics to curb the infection cycle. Significant progress has been made in developing antivirals against flavivirus, with various strategies being explored. However, the lack of specific therapies remains a challenge, necessitating continued research and innovation in this field. In this review article, we reveal viral and host targets indispensable to flavivirus biology and highlight the current landscape of antiviral strategies against flaviviruses, focusing on key compounds, mechanisms, and research findings. This information would help in understanding the development of novel and antiviral drugs.

2. Antivirals Against Flavivirus

Several compounds have been explored to develop antivirals against flavivirus, which will be discussed here extensively. These antivirals are categorized based on different stages of the virus life cycle including entry, replication, polyprotein synthesis and processing, assembly, and virus egress (Figure 1). We also included anti-flavivirus drugs with unidentified targets, artificial microRNAs, and exosomes identified as antivirals (Figure 2).

2.1. Entry Inhibitors

Flaviviruses are enveloped viruses that employ the viral E glycoprotein to enter cells. Flaviviruses accomplish this through binding with attachment factors on cell membranes, such as glycosaminoglycans (GAGs), integrins, and C-type lectin receptors. The low affinity and non-specific nature of this contact, mediated by domain III of envelope proteins (EDIII), ensures adherence of a large number of virions to the cell surface [1]. Receptor-specific binding and clathrin-coated pockets determine the development of endocytic vesicles and the invagination of the plasma membrane. The virus undergoes conformational changes in the endosome, such as the trimerization of the E protein, which is required for membrane fusion [2]. Following capsid protein rearrangements, viral RNA dissociates and releases into the cytoplasm [3,4]. To intervene in the virus entry process, a plethora of viral-E-glycoprotein-targeting drugs were developed, a couple of which are reviewed in detail in the next section (Table 1).
Cell-based high-throughput screening (HTS) identified BP34610, a small-molecule inhibitor that recognizes the N-terminal stem region of the E protein. The compound (EC50 = 0.48 ± 0.06 μM) inhibited replication of DENV and JEV [28], whereas compound 3-110-224.1.1, a synthetic peptide, inhibited DENV (IC90 = 0.77 μM), ZIKV (IC90 of 4.0 ± 0.6 μM), WNV, and JEV (IC90 = 32 μM) by interacting with the envelope protein (E) [20]. Aside from this, computational methods have been used to synthesize peptides targeting protein interfaces involved in viral membrane fusion. This advancement has allowed peptide drugs specifically against the epitope to be designed, with good physiochemical properties and oral bioavailability [44]. In the following sections, we highlight a few crucial inhibitors against flaviviruses.
Z2, a synthetic peptide, is directed against the E protein that is conserved in all flaviviruses. This peptide inhibited infection by ZIKV (IC50 = 1.75 ± 0.13 μM), DENV (IC50 = 4 and 5 μM), and YFV (IC50 = 5 μM) in in vitro assays. Furthermore, Z2 treatment prolonged the survival of ZIKV-infected AG129 mice and reduced the viral load in pregnant ZIKV-infected mice and their developing fetuses. Also, the pups did not exhibit congenital brain development, and the mother did not have a fever or rash, establishing that Z2 is safe for pregnant mice and can be transferred vertically, like ZIKV. The antiviral effectiveness of Z2 can be attributed to its ability to bind with E protein and compromise membrane integrity, hence inactivating virion [5]. Similarly, P5 peptides derived from helix 2 of the JEV E protein stem region demonstrated antiviral efficacy against both JEV (IC50 = 3.93 nM) and ZIKV (IC50 = 3.27 μM) with 50% inhibitory concentration (IC50) in the nanomolar and micromolar range, respectively. P5 peptide acts by blocking the conformational change of E protein induced by acidic pH. Treatment with P5 peptide reduced virus replication and inflammation in the brains of mice infected with JEV. ZIKV-challenged type I and type II interferon-receptor-deficient (AG6) mice had abrogated histological abnormalities in the brain and testes [9]. Moreover, another peptide designated DN59, analogous to the stem domain of DENV, inhibited the replication of both DENV (>99% inhibition of plaque formation at concentrations of <25 microns) and WNV (>99% inhibition at <25 microns) [18,45].
DET2 (IC50 above 500 µM) and DET4 (IC50 of 35 μM) are synthetic 10-mer peptides analogous to EDIII of DENV-2 that exhibited antiviral activities against DENV infection. Transmission electron microscopic (TEM) images confirmed that these peptides impair structural and conformational changes in viral E glycoproteins [10,11], thereby preventing the entry of DENV into the cell. In addition, the dipeptide termed EF potentially reduced DENV viral genome replication in all DENV serotypes. EF interferes with the process of membrane fusion by targeting the hydrophobic pocket of E proteins [12]. DENV-2 treated with 200 μM of EF showed reductions in the intracellular E protein and the viral genome by 83.47% and 84.15%, respectively. RI57, short-chain peptides with 28 amino acids, demonstrated inhibitory activity against DENV serotypes (IC50: DENV-1 = 4.6 ± 3.2 μM, DENV-2 = 7.4 ± 2.8 μM, and DENV-3 = 5.9 ± 4.8 μM) and ZIKV (IC95 = 35 μM). It exerts its effect by inhibiting virus fusion, restricting virus entry into the host cell and subsequent infection [42]. Another peptide, Tat-beclin-1, is an autophagy inducer. Tat-beclin-1 showed antiviral activity against a variety of positive-strand RNA viruses. Tat-beclin-1 D-form (20 mg kg−1 i.p.) improves the clinical outcome, lowers the mortality of WNV-infected mice, and ameliorates neuropathology [46].

2.1.1. Monoclonal Antibodies (mAbs)

Quick implementation of several mAb treatments emerged with the outbreak of the COVID-19 pandemic. mAb treatment has likely saved thousands of lives by avoiding the progression of early disease to life-threatening disorders that would otherwise necessitate hospitalization [47]. The first anti-DENV mAb, Ab513 (EC50 = 200 ng/mL), engineered to target EDIII of DENV-4, efficiently neutralizes all four DENV serotypes [14]. Prophylactic treatment in mouse models infected with DENV-2 substantially lowered viral load and enhanced survival, showing its antiviral effectiveness in the context of heterologous enhancing antibodies. In addition, human monoclonal antibody (HmAb) 2D22 binds to viral envelope (E) proteins in a dimeric form, hindering the E protein rearrangement essential for virus fusion. Similar to Ab513, HmAb 2D22 neutralizes all four DENV serotypes. HMAb 2D22’s epitope could be a vaccination or therapeutic target. Administration of 2D22 (20 µg/200 µL, i.p.) averted death in AG129 mice challenged with a lethal dose of DENV-2, most likely by preventing the antibody-enhanced lethal vascular leakage [14,15,16].
One potential ZIKV medication is human anti-ZIKV immunoglobulin (ZIKV-Ig), which is currently the subject of a phase 1 double-blind, randomized, placebo-controlled trial [17]. Additional therapeutic antibodies, such as TY014, Tyzivumab, and MGAWN1, were created [48] to neutralize WNV, ZIKV, and YFV infections. These three antibodies underwent trial analysis; however, TY014 completed the phase 1 clinical trial successfully, while Tyzivumab was withdrawn from one of two phase 2 clinical studies due to issues with the cohort evaluation caused by an insufficient number of ZIKV-infected patients. Like Tyzivumab, phase 1 data for MGAWN1 showed that this recombinant humanized monoclonal antibody is effectively and safely tolerated in healthy subjects. MGAWN1 had to be dropped, similar to Tyzivumab, due to inadequate enrollment [19].
AC-10 (IC50 < 25 ng/mL), neutralizing monoclonal antibodies isolated from a patient with an active ZIKV infection, effectively neutralized virus infection in Vero cells. It also enhanced internalization of virions into human leukemia K562 cells [43]. ZKA190 (15 mg/kg, i.p.) and FIT-1 (15 mg/kg, i.p.) displayed high neutralizing efficacy and prevented morbidity and mortality of ZIKV-infected mice [21]. Further, B cell epitope mapping studies on ZIKV-infected subjects identified ZIKV-117, a human monoclonal antibody. ZIKV-117 showed strong ZIKV neutralization through its binding affinity towards E protein. Treatment of ZIKV-infected pregnant and non-pregnant mice with ZIKV-117 (100 µg, i.p.) abrogated histopathological changes in the placenta and reduced ZIKV infection and mortality in pregnant mice and their fetus [22]. In contrast, two subsets of E-dimer epitope (EDE) antibodies, EDE1 and EDE2, isolated from a dengue patient, potentially neutralize the serogroups of ZIKV (EDE1/EDE2, IC50 = 0.1 nM) and DENV (EDE1, IC50 = 0.39 nM (DENV-1), 0.24 nM (DENV-2), 0.64 nM (DENV-3), and 1.13 nM (DENV-4); EDE2, IC50 = 0.11 nM (DENV-1), 0.07 nM (DENV-2), 0.11 nM (DENV-3), and 7.79 nM (DENV-4)) [23]. Further, mAb WNV-86 potently neutralized WNV with a 50% inhibitory concentration of 2 ng mL−1. A single dose of WNV-86 following 2 days of WNV infection protected mice from the lethal WNV challenge [24]. Thus, this implies that a structure-based rationale paves the way for future antibody-based therapeutic approaches. Monoclonal antibody therapy has several disadvantages, such as limited availability and accessibility, high cost, the necessity of early intervention, and the risk of side effects [49]. Particularly with mAbs, it is important to take into account the possibility of antibody-dependent enhancement (ADE) exacerbation, as mAbs against RSV, MERS, and SARS-CoV-2 have been demonstrated to induce ADE [50].

2.1.2. Synthetic and Natural Inhibitors

Synthetic agents with broad-spectrum action against numerous flavivirus species are of particular interest. An array of cyanohydrazones were identified to avert DENV, ZIKV, and JEV infection. Particularly, JBJ-01-162-04 (IC90 = 1.5 ± 0.4 μM) suppresses DENV in an in vivo model. Similarly, 2,4-diamino pyrimidines (2-12-2, IC90 = 5 μM; 7-148-6, IC90 = 25 μM) and 4,6-disubstituted pyrimidines (GNF-2, IC90 = 15 μM; 1-100-1, IC90 = 7.5 μM) target the DENV E protein’s prefusion form and impede membrane fusion, preventing viral entry [13]. These molecules bind to a βOG pocket between domains I and II of E protein that appears to be conserved among DENV, WNV, ZIKV, and JEV.
Gossypol, a natural substance, effectively prevented ZIKV infection in nearly all 10 strains. It also demonstrated minimal to no cytotoxicity against four serotypes of DENV human strains. Gossypol (ZIKV: IC50 = 3.48 µM; DENV-1: IC50 = 1.87 µM; DENV-2: IC50 = 1.89 µM; DENV-4: IC50 = 2.6 µM) was found to bind to the ZIKV E protein, most likely EDIII, suggesting that it could identify regions and segments of the ZIKV and DENV E proteins that are highly conserved [25]. Further, the antiviral properties of curcumin have been extensively investigated. It shows antiviral activity against many viruses including chikungunya (CHIKV) (IC50 = 3.89 µM), ZIKV (IC50 = 1.90 µM), and DENV (IC50 = 11.51 µM). Prior to infection, the virus treated with curcumin exhibited decreased viral replication, indicating that curcumin prevents the attachment of viruses to cell surfaces [26,27].
Naturally occurring substances, geraniin and palmatine, showed anti-flaviviral efficacy. Palmatine, a protoberberine alkaloid, derived from Coptis chinensis Franch inhibits WNV (IC50 = 3.6 μM), DENV-2 (IC50 = 26.4 μM), and YFV (IC50 = 7.3 μM). It has been demonstrated that palmatine prevents infection in a variety of cell lines. It exerts its effect by inhibiting NS2B–NS3 protease activity [31]. Thus, this alkaloid could potentially be used to treat flavivirus infections. Further, geraniin prepared from Nephelium lappaceum rind inhibited DENV-2 plaque formation, with an IC50 of 1.75 μM. Geraniin binds to the EDIII protein, thus interfering with the initial cell–virus contact in vitro and in vivo. It has been demonstrated that natural ingredients such as flavonoids and naturally occurring phenols like myricetin (IC50 = 1.3 ± 0.1 μM), quercetin (IC50 = 2.4 ± 0.2 μM), luteolin (IC50 = 2.7 ± 0.3 μM), isorhamnetin (IC50 = 15.5 ± 0.7 μM), apigenin (IC50 = 56.3 ± 0.9 μM), and curcumin (IC50 = 3.5 ± 0.2 μM) block ZIKV NS2B–NS3. According to molecular docking, these compounds bind to a pocket at the rear of the active site of ZIKV NS2B–NS3 and block it by allosterically influencing its structure–activity attribute [51]. Of note, dialkylated flavanones named chartaceous A-F (1–6) (IC50 = 9.0 ± 3.5 μM) isolated from the bark of Cryptocarya chartacea inhibited dengue virus by interrupting NS5 RNA-dependent RNA polymerase (RdRp) activity, serving as potent non-nucleosidic inhibitors of DENV [52].

2.1.3. Host Function Inhibitors

The existing “one drug, one bug” antiviral strategy frequently leads to the evolution of drug resistance and is not readily scalable to meet the unmet clinical need posed by new viruses.
Targeting the host component is an alternative broad-spectrum antiviral approach with the potential to provide a stronger barrier to resistance. This offers the prospect of repurposing safe, previously approved medications that modulate particular host functions (Table 2). The host-targeted strategy is especially appealing for the treatment of new viral infections for that currently have no treatment [36].
Prochlorperazine (PCZ) (EC50 of 88 nM) is a phenothiazine derivative that serves as a dopamine D2 receptor (D2R) antagonist and exhibits antiviral effects against DENV-1, DENV-2, and JEV. A study demonstrated that the drugs inhibit DENV entrance by targeting D2-receptor- and clathrin-associated pathways, thus hampering viral binding and entrance [32]. Moreover, Nawa M. et al. evaluated the effect of chlorpromazine on the entry of JEV. Chlorpromazine blocks the clathrin-mediated endocytic pathway in the case of JEV by accumulating AP-2 and clathrin in endosomal compartments, suppressing the development of clathrin-coated pits. Interestingly, the disruption of clathrin inhibited the replication of JEV (at a dose of 10 μg mL−1) and WNV (at a dose of 5 μg mL−1) [41,53,61].
Sterol regulatory element binding protein (SREBP) regulates cellular lipidomics. SREBP inhibitors, at a dose of 35 μM, including fatostatin, PF-429242, and nordihydroguaiaretic acid (NDGA) and its derivatives, interfered with the SREBP-dependent lipidomic reprogramming and potently suppressed WNV, DENV, and ZIKV infection in vitro [62]. Likewise, Daptomycin (EC50 values between 0.1 and 1.0 µM) is a lipopeptide antibiotic that inhibits ZIKV [33,62].
Nanchangmycin (IC50 ranges from 0.1 to 0.4 μM) produced by Streptomyces nanchangensis effectively inhibits the entry of the ZIKV along with WNV, DENV, and CHIKV [34]. Antiviral activity against WNV, DENV, and ZIKV has been evaluated for FDA-approved anticancer drugs. Sunitinib, a receptor tyrosine kinase antagonist, and erlotinib, an epidermal growth factor tyrosine kinase inhibitor, were investigated in combination against DENV in IFN-α/β and IFN-γ receptor-deficient murine models. Sunitinib (EC50 = 1.962 μM) and erlotinib (EC50 = 4.07 μM) treatment reduced viremia and delayed the onset of paralysis in DENV-infected AG-B6 mice. Also, the combination therapy demonstrated antiviral efficacy in monocyte-derived dendritic cells (MDDCs) [36]. Further, adenosine monophosphate-activated protein kinase (AMPK) agonists PF-06409577, metformin, and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) demonstrated antiviral potentials in WNV (EC50 = 8.2 μM), DENV (EC50 = 4.6 μM), and ZIKV (EC50 = 0.9 μM) infection and inhibited viral replication through the impairment of AMPK phosphorylation [54]. Further, metformin (50 mg/kg/day) treatment enhanced the survival rate of DENV-infected AG129 mice and attenuated severe DENV infection in a cohort study [63,64]. Furthermore, 25-hydroxylcholesterol (25HC) regulates lipid metabolism. It acts by blocking the fusion of the virus and the cell membrane, thus interfering with the internalization of DENV (IC50 = 406 nM), YFV (IC50 = 526 nM), WNV (IC50 = 109 nM), and ZIKV (IC50 = 188 nM), thereby serving as a pan-flavivirus inhibitor. 25HC treatment inhibited ZIKV infection in human cortical tissue in vitro and significantly reduced viremia and mortality in ZIKV-infected BALB/c and AG129 mice. It also demonstrated protection in pregnant mice and alleviated microcephaly in the fetal brain. Moreover, 25HC administration attenuated ZIKV infection and associated clinical signs in rhesus monkeys [37]. Further, PF-05175157 (EC50 = 2.7 ± 1.3 μM) an inhibitor of coenzyme acetyl, reduced viremia and conferred protection against WNV infection in a mouse model [55]. A specific inhibitor called GW4869 (doses ranging from 2 μM to 10 μM), which targets sphingomyelinase-2, demonstrated a reduction in WNV and ZIKV in human fetal astrocytes [56,57].
Endosome acidification is critical for viral genome release into the cytoplasm. Chloroquine (CQ), a 9-aminoquinoline, and its hydroxy-analog, hydroxychloroquine, with a long history of usage as an antimalarial drug, have been studied for the clinical treatment of viral diseases. Treatment with CQ (regimen of 600 mg on day 2 and 300 mg on day 3) raises the trans-Golgi network and lysosome pH, which interferes with endosomal fusion and virus maturation. CQ potentially inhibited ZIKV infectivity in human neural stem cells and primary amnion cells [33,40,57]. CQ (dose of 12 and 16 μM) reduced the DENV replication in THP-1 and human dendritic cells. Further clinical trials conducted on DENV patients lowered the incidence of dengue hemorrhagic fever but failed to alleviate viremia [58]. Niclosamide targets the NS2B-binding site on NS3, which is conserved among flavivirus NS3 proteases, allowing it to have broad anti-flaviviral efficacy. It is highly efficient against DENV (EC50 = 0.55 ± 0.05 μM), WNV (EC50 = 0.54 ± 0.17 μM), YFV (EC50 = 0.84 ± 0.02 μM), ZIKV (EC50 = 0.48 ± 0.06 μM), and JEV (EC50 = 1.02 ± 0.08 μM) [65]. Niclosamide (EC50 = 0.37 μM) acts on multiple targets including endosomal deacidification and disruption of the NS2B–NS3 complex, thereby inhibiting ZIKV replication in human neural progenitor cells and animal models. The observed antiviral activity is likely due to its combinatorial effect on both viral and host components [59].

3. Viral Replication and Polyprotein Synthesis and Processing

The flavivirus genome is translated into a single polyprotein via the host’s ribosomal machinery. Viral and cellular proteases cleave the polyprotein and yield structural and non-structural proteins. The structural proteins capsid (C), precursor membrane (prM/M), and envelope (E) are integrated into the virion, whereas the non-structural (NS) proteins NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5 serve to coordinate the intracellular virus replication, assembly, and modulation of host defense mechanisms [66,67], rendering them promising candidates for the development of therapeutic regimens. Some of the most notable are emphasized in the following subsection (Table 3).

3.1. NS1 Inhibitors

NS1 is a non-structural viral protein that facilitates immune evasion, virion assembly, the viral replication cycle, and viral pathogenicity [75,143,144]. It can be found in oligomeric form, co-localized with dsRNA at the site of viral replication. Furthermore, the soluble form of NS1 activates complement-mediated immune suppression and remodels membrane lipids for organizing the replication complex as it engages in the replication of the virus as well. The glycosylation pattern of NS1 has been reported to be conserved at Asn 130 and Asn 207 among all serotypes of DENV, JEV, and YFV [145,146]. This implies that NS1 is a suitable target for the inhibitor development. Celgosivir, a natural alkaloid castanospermine extracted from the Castanospermum australae, is an α-glucosidase inhibitor. Celgosivir inhibited DENV infection (DENV-1 (EC50 = 0.65 + 0.16 μM), DENV-2 (EC50 = 0.22 + 0.01 μM), DENV-3 (EC50 = 0.68 + 0.02 μM), and DENV-4 (EC50 = 0.31 + 0.12 μM)), mitigated viremia, and prolonged survival by inhibiting glycosylation of prM and E in AG129 mice challenged with DENV [147]. This emphasizes that the glycosylation of NS1 is critical for the virulence of the virus.
The phage display peptide library combined with cutting-edge biopanning techniques identified peptide inhibitors against flavivirus infection. In silico analysis and surface plasmon resonance identified four peptides, i.e., peptide 3, 4, 10, and 11, at a concentration of 10 µM that exhibited strong peptide–protein interactions towards DENV NS1. These peptides differentially inhibited DENV replication in all DENV serotypes in infected Huh-7 cells [68]. Further, using phage display technology, mAbs 3C7 and 4D1 were identified, wherein mAb 3C7 neutralized WNV, while mAb 4D1 neutralized both WNV and JEV [69].
In silico molecular docking screening identified a high affinity of deoxycalyxin-A, a flavonoid, towards ZIKV NS1 [70]. However, in vitro and in vivo research is required to confirm this observation. In addition, α-glycosidase inhibitors like castanospermine (500 μM), deoxynojirimycin (500 μM), and N-nonyl-deoxynojirimycin (20-mg/kg/day, i.p.) decreased NS1 expression and viral production in cells [73,74].

3.2. NS2A Inhibitors

Flavivirus NS2A, a hydrophobic protein, is associated with membranes and has a role in RNA replication. NS2A exhibits significant selectivity in binding to the 3′ untranslated region (UTR) of viral RNA and other replication complex constituents. Moreover, NS2A affects the assembly and secretion of virus particles as well as the host antiviral interferon response [148]. So far, no drugs that target the flaviviral protein NS2A have been studied. However, more work needs to be conducted to understand how NS2 interacts with lipids and evades host immune regulation.

3.3. NS2B–NS3 Inhibitors

NS3 is a highly conserved protein; its structure comprises two domains: a helicase with an NTPase domain at the C-terminus that aids in the replication of viral genome RNA, and a protease with a trypsin-like serine domain at the N-terminus for polyprotein cleavage. It is noteworthy that viral NS2B is a cofactor for NS3’s protease activity. The coupling of NS3 and NS2B forms a complex that is indispensable for polyprotein processing. Given that NS2B–NS3 plays a crucial role in viral replication, it may be a potential target for antiviral drug studies. A plethora of studies conducted in vitro have demonstrated NS2B–NS3-specific inhibitors including peptidomimetic inhibitors, niclosamide (aforementioned), novobiocin (DENV: IC50 = 20,379 nM; ZIKV: IC50 = 17,683 nM), and temoporfin (IC50 = 1.1 μM). Interaction of novobiocin with the ZIKV NS2B–NS3 binding pocket strongly inhibits ZIKV and DENV replication in cell cultures. It also reduced the viral load and enhanced survival in the treated cohort in mouse models [77,78].
HTS has significantly enhanced pharmacological research through swift and effective selection of novel compounds. This approach led to the identification of temoporfin as an NS2B–NS3 inhibitor. Structural docking revealed that temoporfin binds to NS3 pockets that include key NS2B residues, limiting flavivirus polyprotein processing in a non-competitive way. Temoporfin not only suppressed DENV, YFV, WNV, and JEV in vitro but also reduced ZIKV-induced viremia and mortality in mouse models in addition to inhibiting ZIKV replication in human placental and brain progenitor cells [65].
Split luciferase complementation screening identified JMX0207, a niclosamide that intervenes in NS2B–NS3 interactions. JMX0207 effectively inhibited DENV and ZIKV RNA synthesis in A549 cells and human induced pluripotent stem cells, respectively. Also, JMX0207 (40 mg/kg, oral) treatment diminished ZIKV infection in the 3D organoid and A129 ZIKV mouse model [79]. Another example of drug repurposing is Nelfinavir, an FDA-approved drug. It acts as an NS2B–NS3 and NSP2 protease inhibitor and potentially inhibits DENV-2 (EC50 = 3.5 ± 0.4 µM) and CHKV (EC50 = 14 ± 1 µM) infection in virus cell-based assays [80].
Similarly, carbazole amidines compound 17, a bis-amidine derivative of N–Me carbazole (IC50 = 0.52 μM), showed an antagonistic effect against ZIKV NS2B–NS3 protease. Thus, it efficiently suppressed ZIKV expression in vitro [81]. Further, the non-peptide molecules C30H25NO5 (IC50 = 14.9 ± 2.9 μM) and C34H23NO7S2 (IC50 > 11.8 ± 0.2 μM) significantly blocked the DENV NS2B–NS3 protease exhibiting in vitro antiviral activity [82]. Virtual and biochemical screening of NCI library compounds identified NSC135618 (IC50 = 1.8 μM), an allosteric inhibitor that blocked the conformational change of the NS2B–NS3 protease complex and showed antiviral efficacy against all the flaviviruses including DENV, WNV, ZIKV, and YFV in cell culture [83].
HTS assay employed to identify inhibitors of DENV replication revealed that benzoxazole inhibitors such as ST-610 inhibited the DENV-2 viral titer with an EC50 of 0.272 μM and an EC90 value of 3.59 μM. ST-610 inhibits double-stranded RNA’s (dsRNA) unwinding activity via acting on the NS3 helicase and preventing it from attaching to viral RNA. Further, ST-610 can reduce viremia in AG129 mice infected with DENV [97].
Likewise, DENV ATPase HTS assay screening discovered ML283 (IC50 = 500 nM), a benzothiazole oligomer, that prevented NS3h-catalyzed ATP hydrolysis, thus blocking the RNA unwinding process of WNV and DENV NS3 in cell-based assays [99]. Further, SYC-1307 showed antiviral efficacy against multiple flaviviruses (DENV-2 IC50 0.59 μM, DENV-3 IC50 0.52 μM, WNV IC50 0.78 μM, and ZIKV IC50 0.2 μM), suggesting SYC-1307 is a pan-flavivirus protease inhibitor that targets the hydrophobic pocket of NS3, catalytically inactivating the conformation, which as a result significantly improved the survival of ZIKV-infected mice [98].
Another study by Aleshin et al. provides evidence for the differential substrate selectivity of DENV and WNV proteases. According to structural analysis, aprotinin, also referred to as bovine pancreatic trypsin inhibitor (BPTI), binds to the active site of NS2B–NS3pro and forms an inactive conformation of the oxyanion hole, through an induced-fit mechanism. This complex oxyanion hole is a catalytically competent and stable conformation that perturbs flaviviral proteases in vitro [86]. Further, screening of antimicrobial compounds and in silico docking analysis identified the benzimidazole compound MB21 as a potential inhibitor of NS2B–NS3 of DENV. The benzimidazole moiety of MB21 (IC50  =  5.95 μM) interacts with an allosteric site (Ala125) on the protease that potentially inhibits replication of all the DENV serotypes in vitro with an IC50 of 5.9 μM [85].
The screening of a natural compound library and octet binding assay using biotinylated ZIKV pro showed the potential for ZP10 (theaflavin-3,3′-digallate) as ZIKV pro inhibitors. ZP10 (IC50 = 2.3 μM) mitigated the viral RNA copy numbers and expression of ZIKV envelope proteins in vitro. The antiviral efficacy of ZP10 has been ascribed to the inhibition of ZIKV polyprotein cleavage by specifically targeting the ZIKV protease [84].
Screening of allosteric exosite-targeting inhibitors coupled with structural modeling revealed NSC157058 as a homolog of NS2B–NS3pro that likely intervenes with the binding of the NS2B cofactor relative to the NS3pro active site, thereby inactivating the protease activity. Incubation of ZIKV-infected human fetal neural and SJL mice with NSC157058 (30 mg/kg) reduced ZIKV infection and viremia [101]. Further, the same strategy indicated that hydroxychloroquine (HCQ), a chloroquine derivative and FDA-approved drug, binds to NS2B–NS3 protease and fits in the active site pocket as well as demonstrating suppression of the number of foci in ZIKV-infected trophoblasts with HCQ treatment with an inhibition constant (Ki) of 92.34 ± 11.91 μM [87].
Interestingly, based on split luciferase complementation (SLC), an FDA-approved drug, methylene blue (MB) and Erythrosin B were identified and characterized to have an inhibitory effect against the viral NS2B–NS3 protease by limiting polyprotein processing, overall leading to a reduced level of viral RNA synthesis and viral protein expression in cell-based assays. The EC50 values for ZIKV and DENV-2 are in the low micromolar and nanomolar range in cell-based antiviral assays. In addition, MB (IC50 = 29 μM) and Erythrosin B (EC50 = 0.3 μM) treatment conferred protection against ZIKV in 3D mini-brain organoids and prolonged the survival of A129 mice infected with ZIKV [89,90].
An in silico docking library search revealed in vitro anti-helicase activity for ivermectin. Ivermectin attenuated the in vitro replication of different flaviviruses [YFV (IC50 = 0.12 ± 0.01), DENV (IC50 = 0.50 ± 0.07), and WNV (IC50 = 0.35 ± 0.04)] by impairing the dsRNA unwinding activity with IC50 values in the sub-micromolar range. However, ivermectin failed to protect AG129 mice infected with ZIKV [92,93]. Similarly, HTS identified anti-NS3 helicase activity of suramin. CPE reduction assays demonstrated anti-ZIKV and anti-DENV activity of suramin with an IC50 value in the micromolar range. Suramin treatment in vitro caused reduction in ZIKV and DENV genome replication [100,149]. Further, screening of inhibitors that target the NS2B3 protease resulted in the characterization of bortezomib (EC50 = 8 nM), which effectively inhibited ZIKV and DENV replication in vitro. Bortezomib treatment ubiquitinates NS2AB3 protease and perturbs its self-cleavage that substantially impaired virus replication [95]. An integrated anchor-based screening approach for screening of the FDA drug dataset against DENV NS3 protease found that asunaprevir (EC50 = 10.4 μM) and telaprevir (EC50 = 24.5 μM) efficiently lowered viral plaque count in vitro, confirming strong anti-DENV activity [96].

3.4. NS4A and NS4B Inhibitors

ER-derived cytoplasmic vesicular packets (VPs) or convoluted membranes (CMs) act as scaffolds for the viral replication complex (VRC). NS4A is one of the key elements of the VRCs, including other NS proteins and viral dsRNA. The N-terminal of NS4A is composed of an amphipathic α-helix, which modulates endoplasmic reticulum (ER) membrane curvature, while the C-terminal of NS4A has a 2k fragment that serves as a signal peptide for the ER localization of NS4B. NS4A mediates anchoring of VRCs to the perinuclear membrane, allowing efficient viral RNA replication. Flavivirus NS4A functions as a cofactor and regulates the ATPase activity of NS3 helicase. In addition, NS4A contributes to the pathogenesis of flaviviruses by antagonizing IFN-I production and RLR signaling, altering the UPR and autophagy as well as hijacking several cellular signaling pathways, resulting in developmental defects. Hence, it represents a promising antiviral drug target [150,151]. Of note, in a cell-line-based model, functional investigation of SBI-0090799 (IC50 = 2.1 μM) and Compound B (IC50 = 1.32–4.12 μM) showed that they prevented ER membrane remodeling by inhibiting ZIKV and DENV’s NS4A activity, respectively. Thus, they represent a promising lead candidate for further development as an antiviral [102,109].
NS4B is the largest non-structural protein of flavivirus. It shares functional similarities with NS4A. NS4B recruits the host protein to remodel the membrane topology of ER. NS4B interacts with other viral proteins, including NS1, NS2B, NS3, and NS4A, facilitating the formation of a viral replication complex. It pairs with NS3 and enhances the helicase activity of NS3. Also, the interaction with chaperones promotes optimal polyprotein folding and expression. Moreover, NS4B aids in host immune evasion by RNAi pathway inhibition, autophagy and apoptosis modulation, and type I interferon signaling mitigation [152,153]. Because of all these attributes, NS4B is an appealing target for antiviral research. Research leveraging compounds targeting NS4B activity has discovered quite a few compounds with potent antiviral activity.
In particular, JNJ-A07 and JNJ-1802 prohibited the formation of the NS3–NS4B complex. JNJ-A07 induces a conformational shift in the cytosolic loop of NS4B by blocking the NS4B–NS3 interaction. JNJ-A07 is a pan-serotype DENV antiviral since it can also affect a wide range of DENV isolates at nanomolar to picomolar concentrations. Further, JNJ-A07 (10 mg per kg) treatment reduced viremia in DENV-infected AG129 mice. An analog of JNJ-A07 (JNJ-64281802) demonstrated favorable pharmacokinetic and safety profile, thus was registered for a phase 2 randomized, double-blind, placebo-controlled clinical trial to examine therapeutic potential against DENV infections in humans. However, no publication of the trial’s findings has been made [103].
JNJ-1802, a small-molecule inhibitor belonging to the same chemical series as JNJ-A07, showed antiviral activity in picomolar to low nanomolar concentrations against all DENV serotypes, JEV, WNV, and ZIKV in vitro with mean EC50 values ranging from 0.25 µM to 1.1 µM. JNJ-1802 dosing alleviated the viral RNA load and conferred protection in AG129 mice challenged with DENV serotypes (DENV-1, DENV-3, and DENV-4) as well as in monkeys infected with DENV-2 [154]. JNJ-1802 exhibited excellent safety, tolerability, and pharmacokinetics; thus, it successfully completed the phase 1 clinical trial in healthy human participants (NCT05201794) and in patients with confirmed DENV infection (NCT04906980) [104,155].
Investigation of the structure–activity association found that JMX0254 (100 mg/kg, oral) and BDAA (IC50 = 0.17 μM) target NS4 activity and inhibited expression of DENV in AG129 mice and YFV, respectively [107,108]. Another compound, SBI-0090799 (2E)-N-benzyl-3-(4-butoxyphenyl) prop-2-enamide) (IC50 = 2.1 μM), obstructs the formation of the membrane replication complex by blocking NS4, which suppressed the replication of several ZIKV strains in a cell-based assay [109].
It has been reported that NITD-688 antagonizes NS4B viral protein. However, the specific mechanism is being studied. Cell-based CPE assay verified that NITD-688 was potent against all DENV serotypes. Nuclear magnetic resonance confirmed the direct interaction of NITD-688 with NS4B, as NITD-688 failed to bind with mutated NS4B at T215A and A222V. NITD-688 (30 mg per kg, oral) alleviated viremia in DENV-infected AG129 mice. Further, it showed good in vivo pharmacokinetic properties and oral bioavailability post-administration in mice, rats, and dogs. Overall, the findings validate NITD-688 as a preclinical candidate for DENV therapy [105].
Screening of the FDA-approved drugs library identified manidipine (at a 10 μM concentration) to be efficient against all flaviviruses including JEV, DENV, ZIKV, and WNV in vitro. It prevented brain damage in JEV-infected rats but did not affect viremia. Manidipine substantially lowered viral RNA synthesis by targeting NS4B, but the mechanism of its antiviral activity remains unclear [106]. Lycorine, an alkaloid, attenuated the viral titers of WNV, DENV, and YFV at a concentration of 1.2 microns and potently inhibited flaviviruses in a cell-based assay. Moreover, AZD0530 and dasatinib exhibit substantial efficacy in inhibiting DENV-2 replication in cell culture. NS4B inhibition is the mediating mechanism for these drugs, although their precise processes are yet to be determined [110].

3.5. NS5 Inhibitors

The biggest flavivirus protein, NS5 harbors both methyltransferase (MTase) at the N terminal and RdRp at C terminal, playing a vital role in RNA replication. As a result, it has been researched the most for the development of antiviral drugs against flavivirus [156]. There are several types of antiviral drugs that target NS5, including MTase inhibitors, non-nucleoside inhibitors, and nucleoside inhibitors. Drugs have been screened and repurposed to identify nucleoside analogs that target viral polymerases and exhibit flavivirus-opposing action both in vitro and in vivo [157]. A plethora of nucleoside analogs with high antiviral potency and favorable pharmacokinetics have already received FDA approval for use as antiviral medication to treat HIV infection [158,159,160], HCV [161], and herpesvirus [162].

3.5.1. Nucleoside Analogs

The most appealing drugs are usually nucleoside/nucleotide inhibitors (NIs) as they target conserved RdRp active sites prone to relatively low mutation rates in these regions, allowing broad-spectrum activity and a strong barrier to resistance. Structurally, NIs are remarkably similar, but a few small variations in heterobase moiety, kinds of glycosidic linkages, and substitutions at various sugar positions can greatly impact their pharmacokinetic (PK) and active characteristics. Their antiviral activity stems from the replication-induced premature termination of RNA genome synthesis.
An adenosine analog known as BCX4430 (Galidesivir) shows broad-spectrum antiviral activity against Ebola virus (IC50 = 11.8 μM), Marburg virus (IC50 = 5.0 μM), YFV (IC50 = 14.1 μM), JEV (IC50 = 43.6 μM), and DENV-2 (IC50 = 32.8 μM) both in cell based assays and in mouse models by inhibiting viral RNA polymerase by non-obligate RNA chain termination [112,113]. Phase 1 trials for this drug are currently underway, but the results are not yet available. Furthermore, NITD008, another adenosine analog, showed an inhibitory effect on the infection of DENV-2 (EC50 = 0.64 μM), WNV, YFV, ZIKV (EC50 = 241 nM), and hepatitis C virus (HCV) (EC50 = 0.11 μM) in animal models and cell lines. Phosphorylated NITD008 acts as a chain terminator and directly inhibits RdRp during the synthesis of viral RNA replication, preventing mortality in DENV- and ZIKV-infected mice [118,163]. Similarly, favipiravir, a pyrazinecarboxamide derivative, showed a good tolerance profile in patients and is currently undergoing clinical studies to treat SARS-CoV-2 [164], Ebola virus [165], and influenza virus. Favipiravir (EC50 = 61.88 μM) not only conferred protection in mice from meningitis and encephalitis caused by a lethal dose of YFV and WNV [115], but was also successful against ZIKV, indicated through in vitro study, which suggested an increase in the frequency of mutations in the viral genome, leading to generation of non-infectious defective viral particles and thus declining viral infectivity in subsequent progeny [166]. Favipiravir (EC50: from 62 to >500 μM) treatment was well tolerated and demonstrated a statistically significant decline in the ZIKV virus load in the plasma of cynomolgus macaques [116]. Monophosphate prodrug analogs of 2′-deoxy-2′-fluoro-2′-C-methylguanosine are strong inhibitors of the RdRp of the HCV and exhibit anti-DENV properties in a cell-based assay. In an AG129 mouse model, cyclic phosphoramidite, prodrug 17, and compound 17 (100 mg/kg) showed decreases in viremia. Nevertheless, the compound failed to attain the “no observed adverse effect level” (NOAEL) in dogs due to mild pulmonary inflammation and hemorrhage. The drug did not progress to the clinical stage because of the severity noted in the preclinical safety assessment [167].
Prodrug balapiravir (R1479) is a cytidine analog that was used in the clinical trial for the treatment of patients infected with chronic HCV, resulting in an improved outcome for patients [168]. In contrast, balapiravir (3000 mg, oral) administration in DENV patients did not differ from drug and placebo treatment in terms of antiviral response, cytokine profile, or time to fever [169]. Likewise, in vitro and in vivo testing of NITD-008, an adenosine nucleoside analog inhibitor, demonstrated strong antiviral efficacy against ZIKV (EC50 = 241 nM), TBEV (EC50 = 3.31 μM), and DENV (EC50 = 0.64 μM) [118,170]. However, preclinical toxicity, including weight loss, decreased motor activity, bloody feces, irreversible corneal opacities, and blood anomaly, prevented the drug from moving forward with clinical trials [163].
The guanosine nucleotide analog prodrug AT-752 has been shown to inhibit RdRp activity of NS5 protein. AT-752 exhibited strong in vitro action against DENV-2 (EC50 = 0.48 μM), DENV-3 (EC50 = 0.77 μM), WNV (EC50 = 1.41 μM), YFV (EC50 = 0.31 μM), ZIKV (EC50 = 0.64 μM), and JEV (EC50 = 0.21 μM). In addition, an AG129 mouse model of DENV infection and hamsters infected with YFV showed a reduction in viremia, improved health score, and prolonged survival with AT-752 administration. Preclinical toxicology studies in mice, rats, and monkeys have indicated no adverse clinical signs. Thus, AT-752 has successfully finished its phase 1 clinical trial (NCT04722627) and is currently in phase 2. Further, the metabolism of AT-752 yields AT9010, an active triphosphate metabolite that prevails stably in cynomolgus monkey and human plasma. During RNA synthesis, AT-9010 competes with GTP for incorporation, causing chain termination, thus efficiently inhibiting RNA synthesis in vitro and in vivo in a mouse model of DENV infection. The antiviral activity of AT9010 is presently being assessed in a phase 1 experiment [120,121].
In vitro testing of another nucleoside analog, 7DMA (7-deaza-2′-C-methyladenosine), also known as MK-608 (EC50 = ranging from 5 to 15 μM), effectively halted RNA synthesis of flaviviruses such as TBEV, ZIKV, WNV, and DENV by chain termination of nascent RNA. In vivo, 7DMA (50 mg/kg/day) treatment reduced viral titer along with the aversion of neuroinflammation and disease progression in ZIKV-infected AG129 as well as WNV-infected BALB/c mice. Furthermore, administration of 7DMA during infection resulted in 100% survival of WNV-infected mice, while treatment after three days of infection, at the peak of viremia, led to a 90% survival rate, confirming its antiviral activity [122,123,171].
The quest for a pan-flaviviridae drug may benefit from the highly conserved structural and functional homology of RdRp enzymes across different flaviviruses [172]. In a similar vein, sofosbuvir is a clinically approved small-molecule inhibitor against hepatitis C infection. Three-dimensional modeling revealed that the drug pairs with the palm and finger region of RNA polymerase, where incoming nucleotides are integrated into a nascent strand of RNA, thus impairing RdRp activity. Sofosbuvir has shown in vitro antiviral activity against ZIKV (IC50 = 13.6 μM), DENV (IC50 = 8.31 μM), WNV (IC50 = 11.1 ± 4.6 μM), and YFV (IC50 = 4.8 ± 0.2 μM). Notably, sofosbuvir mitigated ZIKV replication and blocked cell death in neural stem cells (NSCs), macrophages/microglia, and brain organoids. Interestingly, pre-treatment and post-treatment of YFV-infected Swiss and A129−/− mice with sofosbuvir (20 mg/kg/day) abrogated hepatic histopathological lesions, weight loss, viral load in the brain, and mortality. Outcomes of YFV-challenged mice were superior with pre-treatment of sofosbuvir than post-treatment [124,125,126,173,174,175].
Ribavirin was originally prescribed to treat HCV and HBV, much like other nucleoside analogs including AT-752, 7DMA, and sofosbuvir. Functionally it acts as an RNA polymerase inhibitor; alleviates RNA replication of DENV, JEV, and ZIKV; and halts cell apoptosis in vitro. Ribavirin (15 mg, i.p.) administration to ZIKV-infected STAT1-deficient mice exhibited prolonged survival, but mice continued to have low viremia levels until death [130,176] as well as failing to inhibit DENV infection in the AG129 mouse model Also, oral ribavirin administration could not rescue JE patients from mortality [123]. However, rifapentine (IC50 = 0.5259 μM), a rifamycin antibiotic, is a potent YFV inhibitor. It interferes with viral attachment and impedes RdRp activity, as shown by binding assays and molecular docking, respectively. Rifapentine increases survival rates, mitigates clinical symptoms, and lowers viral load in both YFV-infected type I interferon receptor knockout A129−/− and wild-type C57BL/6 mice. In fact, it surpasses sofosbuvir, a previously described YFV inhibitor in mice [6]. Further, T-1106, a pyrazine nucleoside analog, showed considerably enhanced survival rates in YFV-infected hamsters. Even low doses of 32 mg/kg body weight per day (120 μmol/kg/day) were effective against the virus, indicating its potential use in the treatment of YFV [133].

3.5.2. Non-Nucleoside Inhibitors

This class of compounds obstructs RdRp conformation change during RNA polymerization by direct interaction with the thumb/palm interface of the RdRp. Given that the X-ray crystallography showed compounds such as N-sulfonylanthranilic acid derivative NITD-1 (IC50 = 7.2 μM), NITD-2 (IC50 = 0.7 μM), NITD-640 (IC50 = 0.9 to 13  μM), NITD-434 (IC50 = from 6 to 17  μM), and NITD107 (IC50 = 113 μM), either pair with the RdRp fingers or that targets the junction between the fingers and palm subdomains exhibits anti-DENV activity against all DENV serotypes in vitro [127,177,178]. Structural activity relationship studies revealed that FDA-approved drugs—efavirenz (IC50 = 25.78 μM), tipranavir (IC50 = 29.85 μM), and dasabuvir (IC50 =  16.12 μM)—halt RNA polymerization by premature termination of RNA synthesis of flaviviruses including WNV, ZIKV, and TBEV in cell culture [129]. Further, lycorine, an alkaloid derived from benzyl phenethylamine, was used both in vitro and in vivo to treat ZIKV infection. The treatment (lycorine—5 mg/kg) lowered the virus replication in the CNS, liver, and serum as well as reduced histopathological lesions in the brain and liver of ZIKV-infected AG6 mice. These mice also showed decreased levels of inflammation. Lycorine directly interacts with finger domains of RdRp that potently inhibit RdRp activity and viral replication [135]. Likewise, molecular docking revealed the binding affinity of TBP (IC50 = 94 nM) towards the residues in the catalytically active site of RdRp. This likely explains the inhibition of ZIKV replication in vitro as well as the viral load in the plasma of immunocompetent BABL/c mice infected with ZIKV [137]. Similarly, emetine (IC50 = 52.9 nM) blocks NS5 function and accumulates in cellular lysosomes with impaired autophagy and lysosomal function, interrupting cellular trafficking of nascent virion and eventually eliminating viral infection in vitro. Further, emetine treatment significantly decreased NS1 and ZIKV viral load in the serum and liver of infected immunocompromised Ifnar1−/− and immunocompetent SJL mice, indicating anti-ZIKV activity [136].
Notably, NS5 harbors nuclear localization sequences (NLSs) which directly interact with importin β1. This interaction is essential for the translocation of the importin-NLS-containing cargo protein complex through the nuclear pore complex (NPC) into the nucleus [179]. Ivermectin (LD50 = 150 μM) has been used as a broad-spectrum inhibitor of importin α/β, which effectively prevented DENV replication in vitro. Pharmacokinetic and pharmacodynamic examination of DENV patients dosed with ivermectin suppressed NS1 expression and alleviated dengue disease severity. Indicating potent antiviral activity of ivermectin towards DENV [131].
Of note, AR-12 (also known as OSU-03012) and its derivatives P12-23 and P12-34a act as inhibitors of the de novo pyrimidine biosynthesis pathway via inhibiting mitochondrial enzyme dihydroorotate dehydrogenase (DHODH) activity, leading to the depletion of uridine monophosphate pool, essentially required for the viral RNA synthesis and protein glycosylation. Thus, they effectively impaired RNA synthesis of DENV (IC50 = 20.3 ± 0.2 µM), ZIKV (IC50 = 21.4 ± 0.2 µM), and JEV (IC50 = 21.4 ± 0.2 µM) in vitro and STAT1-deficient mice infected with DENV [60].

3.5.3. MTase Inhibitors

MTases located at the N termini of NS5 perform dual N7 and 2′-O methyltransferase activities, and guanylyltransferase (GTase) is implicated in RNA cap formation. Extensive research studies into MTase inhibitors revealed that sinefungin, an analog of S-adenosyl-L-methionine (SAM), showed greater affinity for the SAM binding pocket of viral NS5, thus attenuating (IC50 = 0.03 mM) N-7 and (IC50 =  0.041 mM) 2′-O methylation by MTase. This likely explains the inhibited replication of both WNV and DENV [138,139]. Flexible nucleoside analogs known as “fleximers” have shown antiviral effectiveness against a variety of viruses, including the Middle East Respiratory Syndrome coronavirus (MERS-CoV), Ebola virus, and lately, flaviviruses such as ZIKV, DENV, and YFV cell-based systems [141,142]. Since non-structural proteins are substantially conserved in structure and are essential for viral replication, they are vital candidates for therapeutic targeting. As a proof of concept, pan-flavivirus antiviral is conceivable due to the structural homology of indispensable viral non-structural proteins. Consequently, the development of pharmacological inhibitors based on NS-protein-centric research targeting NS4B, NS5, and NS3 efficiently halted virus biogenesis.

4. Assembly and Egress Inhibitors

All flaviviruses are enveloped viruses employing lipid remodeling of their hosts to change the membrane’s fluidity to assemble itself and provide energy for the reproduction of their genomes [180,181,182]. In the ER, the virus first assembles as an immature particle. It is then carried to the trans-Golgi network (TGN), where it matures in response to pH. The host protease furin cleaves the pr domain of prM during the maturation phase, eventually releasing a fully matured infectious virus particle from the cell [183].
Glycosylation of viral proteins is integral for the release of enveloped viruses and infectivity. (Glc)3(Man)9(GlcNAc)2 is a 14-residue oligosaccharide, added to specific asparagine residues on the prM, E proteins, and NS1 of the newly synthesized polypeptide in the ER [184,185], followed by the trimming of terminal glucose by ER Glucosidase I on high-mannose carbohydrates to generate monoglucosylated glycoproteins that further associate with ER chaperones like calnexin and calreticulin for proper folding [186]. Given the functional implication of glycosylation in the virus life cycle, inhibitors of α-glucosidase have shown inhibitory potency against viruses. Iminosugars such as castanospermine (CST) [187] and deoxynojirimycin (DNJ) [74] hinder the removal of the terminal glucose residue on N-linked oligosaccharides of DENV envelope glycoproteins and NS1, leading to the formation of misfolded proteins and defective viral progeny in vitro and in vivo. Further, celgosivir, also known as 6-O-butanoyl castanospermine (DENV-1–4, EC50 =  0.65 + 0.16 μM, 0.22 + 0.01 μM, 0.68 + 0.02 μM, 0.31 + 0.12 μM), demonstrated 100% protective efficacy against lethal infection in DENV-infected AG129 mice [188]. In response to the promising preclinical pharmacology data, a phase 1b randomized, double-blind, placebo-controlled trial of celgosivir was conducted in patients with DENV. However, celgosivir failed to reduce fever or viremia in patients with DENV infection [189]. In addition, no serious adverse events were reported in the phase 1 study of randomized single oral dose safety of UV-4 hydrochloride (UV-4B) [190]. Other promising iminosugars and their derivatives, such as UV-12 (EC50 =  21.71 μM), CM-9-78 (EC50 =  0.42 μM), and CM-10-18 (EC50 =  0.46 μM), demonstrated anti-DENV activity by inhibiting α-glucosidase in vitro and in vivo, so they merit further investigation for application in therapeutic settings [191,192,193].
Flaviviruses modulate the host fatty acid biosynthetic route to alter the lipid composition for nascent virion biogenesis. Rendering pharmacological manipulation of cellular lipids is a desirable antiviral approach. Host-directed antiviral interventions unveiled acetyl-coenzyme A carboxylase (ACC) 1 and 2 inhibitors, such as PF-05175157 [(WNV, EC50 =  2.7 ± 1.3 μM), (ZIKV, EC50 < 1.2 μM), (DENV, EC50 = 1.0 ± 0.3 μM)], TOFA (5-tetradecyloxy-2-furoic acid) (10 μg/mL), and MEDICA 16 (3,3,14,14-tetramethylhexadecanedioic acid) (50 μg/mL), exhibited potency against various flaviviruses, including WNV, DENV, and ZIKV in vitro. as Additionally, PF-05175157 administration significantly decreased viral load in the plasma, brain, liver, kidneys, and lungs of WNV-challenged mice [55,194].
Unlike other host-directed antivirals, SFV785 treatment (at a concentration of 10 μM) could not inhibit viral RNA synthesis or translation within the ER compartment. However, it perturbed the co-localization of the structural E protein with the replication complexes during assembly, which strongly lowered the yield of infectious virions of DENV and YFV [195,196].
Preclinical evaluation of lovastatin (200 mg/kg, oral) using animal models displayed anti-DENV activity. This could be due to a decrease in the cholesterol-rich microdomains and isoprenylated proteins necessary for virus trafficking, thus limiting the entry of the virus into the cell and viremia in vivo. Yet, it failed in a randomized, double-blind, placebo-controlled trial for DENV patients [197,198]. Similarly, ezetimibe (IC50 = 19.15 µM) and atorvastatin (IC50 = 24.12 µM) showed significant antiviral effects in AG129 mice infected with DENV-2 by targeting the cholesterol biosynthetic pathway [197].
The assembly of viral glycoprotein and capsid is a well-coordinated and highly regulated process that relies on cellular signals and the environment to initiate conformational or allosteric changes in the assembly process. By interfering with the dynamics of viral maturation, assembly, and egress, it is possible to perturb virus propagation (Table 4).

C Protein Inhibitors

C protein is the structural protein that interacts with the RNA genome and prM and E proteins before packaging. The aggregation of membrane-associated capsid into the nucleocapsid structure induces the formation of immature virus particles. The mature C is a highly basic protein of 12 kDa that forms homodimers in solution [205], while the residues at the N-terminal region of the capsid are implicated in RNA binding and viral particle formation. Despite being the least conserved of the flavivirus proteins, the capsid’s structural characteristics and charge distribution are both well conserved [206]. Extensive structure–activity relationship (SAR) analysis identified an analog VGTI-A3–03 with anti-DENV-2 potency by promoting the formation and release of noninfectious virions. Further analysis revealed that VGTI-A3–03 (IC90 = 25 nM) directly pairs with C dimers in viral particles and induces the formation of malformed DENV virions in vitro [202]. Likewise, structural, biochemical, and virologic approaches demonstrated that ST148 (EC50 = 0.016 ± 0.01 μM) through hydrophobic interaction forms a complex with C tetramer, which then incorporates into a virion, inducing the production of progeny with defective nucleocapsid uncoating. ST-148 (50 mg/kg/day) administration mitigated the viremia and viral load in the spleen and liver of DENV-challenged AG129 mice. Hence, it showed promising anti-DENV efficacy against all four serotypes in the cell culture and mouse model [203,204]. C protein binds with viral RNA to regulate encapsidation, as well as bind and sequester short interfering RNAs to protect the flavivirus genome from siRNA pathways. Consequently, it presents a desirable target for anti-flaviviruses [207].

5. Anti-Flavivirus Drugs with Unidentified Target

Several drugs have demonstrated antiviral effects against one or more flaviviruses both in vitro and in vivo. For some, though, the course of action is still unclear. These medications include azithromycin (EC50 = 2 to 3 μM), nitazoxanide (EC50 = 0.12 ± 0.04 μg/μM), amodiaquine (EC50 = 1.08 ± 0.09 μM), lanatoside C (IC50 = 0.19 μM), bromocriptine (EC50 = 0.8–1.6 μM), and hippeastrine hydrobromide (HH) (IC50 = 1.95 μM) [208,209,210,211]. As with lanatoside C, its low therapeutical index dose makes it unsuitable, whereas HH was successful in controlling infection in human fetal-like forebrain organoid cultures. In a rat model, ZIKV infection was efficiently controlled with a notable reduction in ZIKV RNA in the brain and ZIKV-induced cellular death. FDA-approved fluoroquinolones enoxacin (EC50 = 18.1 μM), difloxacin (EC50 = 25.4 μM), and ciprofloxacin (EC50 = 56.8 μM) inhibited the replication of the flaviviruses Modoc (MODV), Langat (LGTV), DENV, and ZIKV in HEK-293 cells at micromolar concentrations. ZIKV titer was significantly reduced in mice administered enoxacin, 2.5 times lower than the control [212]. Arbidol has antiviral effects at the micromolar level (EC50 values ranging from 10.57 ± 0.74 to 19.16 ± 0.29 µM) in Vero cells infected with tick-borne encephalitis virus, ZIKV, and WNV [213].
NGI-1 (EC50 = 0.85 μM) demonstrated pan-flavivirus antiviral activity and blocked viral RNA replication of DENV and ZIKV in multiple disease-relevant cell lines [214]. Trametinib is the safest and is efficacious against all of the viruses, inhibiting the replication of ZIKV (EC50 = 3.03 μM) (EC50 = 7.91 μM) and YFV 1000-fold, and DENV-2/3 (EC50 = 2.46/6.33 μM) nearly 100-fold. This pan-antiviral effect indicates that trametinib could be repurposed for the treatment of flaviviral infection [215]. Quinestrol (ZIKV, IC50 = 11.4 μM; DENV-2, IC50 = 15.3 μM; WNV-KUNV, IC50 = 10.1 μM) and raloxifene hydrochloride (ZIKV, IC50 = 7.4 μM; DENV-2, IC50 = 9.3 μM; WNV-KUNV, IC50 = 7.7 μM) are two estrogen receptor modulators that demonstrated minimal cytotoxicity and effective inhibition of ZIKV, DENV, and WNV (Kunjin strain) infection at low micromolar concentrations [216]. Anisomycin, an alkaloid, inhibited DENV (EC50 = 31.3 ± 1.2 nM) and ZIKV (EC50 = 15.9 ± 1.1 nM). A mouse model of ZIKV treated with a modest dose of anisomycin showed a significant decrease in morbidity and mortality [217]. Fenretinide (EC90 = 2.0 μM), also known as 4-hydroxyphenyl retinamide (4-HPR), was shown to be an inhibitor of DENV in cell culture. In an oral DENV infection model in mice, 4-HPR prevents the steady-state build-up of viral genomic RNA and lowers viremia [218]. Furthermore, the antimalarial medication amodiaquine (IC50 = 2.28 μM) conferred protection in hNPCs and a SCID-beige mouse challenged with ZIKV [219].

6. Artificial microRNAs

The discovery of artificial microRNAs (amiRNAs) directed against viral genomes and the antiviral effect of human miRNAs has abrogated neurovirulence and flavivirus infection (Table 5) [220,221,222]. amiRNAs targeting viral conserved regions at 3′UTR were a useful approach for improvements of nucleic acid inhibitors against JEV. Transient expression of two amiRNAs (amiRNA #1 and amiRNA #2) showed a reduction in intracellular viral RNA and non-structural 1 (NS1) protein and decreased the release of infectious viral particles up to 95% quantified by viral plaque assay [223]. In a similar vein, amiRNA targeting a conserved region of gene NS5 significantly reduced the WNV replication as determined by plaque assay, real-time RT-PCR, and Western blot analysis [224]. Further, amiRNA DENV-128 was able to decrease DENV replication [225]. Additionally, short hairpin RNA (shRNA) and small interfering RNA (siRNA) molecules specific to NS2A demonstrated antiviral properties against JEV in vitro [226,227].
To address the limitations of traditional vaccines, novel vaccine candidates, and genetically engineered flaviviruses with MREs that allow host miRNAs to target viral transcription via endogenous RNA silencing have been proposed. In vivo, the MRE-based WNV vaccine including insertion of several MREs of miR-124a with perfect complementarity in tandem flanking regions of the UTR exhibited safe tolerance and persistent expression. Vaccine construct suppressed WNV infection and produced neutralizing antibody titers that protected animals from lethal challenge [33,229]. This study offers proof in favor of employing miRNAs as an antiviral treatment to combat viruses.

7. Exosomes

Exosomes (EVs) or microvesicles released from cells carry viral genomes, proteins, mRNAs, DNA, and miRNAs to target cells, altering the physiological state of the recipient cell. With miRNAs as one of its cargos, EVs can both promote and inhibit viral proliferation; thus, they are the potent players in viral pathogenesis (Table 6) [232,233,234,235]. Viral RNA, DENV-3 290 or DENV-3 5532, was identified in the EVs isolated from DENV-infected human MDDCs. This indicates that DENV utilizes the EV route to differentially modulate immunological response. Tomohiro et al. demonstrated that cell-to-cell transfer of JEV- and DENV-derived sub-genomic replicons contributed to viral propagation and pathogenesis [236].
Of note, EVs derived from PBMCs treated with IFN alpha blocked DENV replication (DENV-3-5532) in recipient cells [237], whereas berbamine treatment decreased the low-density lipoprotein receptor (LDLR) expression at the plasma membrane, which in turn significantly lowered the susceptibility of cells to JEV infection [240]. Interestingly, EVs derived from human saliva showed anti-ZIKV activity [241], while cow- and goat-milk-derived EVs showed anti-DENV activity [245]. Additionally, in EVs overexpressing Defensin alpha 1B (DEFA1), interferon-induced transmembrane protein 3 (IFITM3), and loaded with LL-37, cathelicidin antimicrobial peptide exerts significant anti-ZIKV activity [242,243,244]. Further, enrichment of EVs isolated from cells either infected with WNV and/or stimulated by IFN-alpha revealed differential expression of mRNAs and miRNAs. This suggests that WNV infection modulates the RNA cargo composition in EVs through both IFN-independent and IFN-dependent pathways [238]. Of note, the ZIKV study employing the rhesus macaque trophoblast stem cell model identified EV-derived diagnostic non-invasive miRNA markers that may aid in the diagnosis of placental infection [239]. Unlike other pharmacological molecules, EVs can pass the blood–brain barrier, have low immunogenicity, and are stable in biofluids, rendering it a promising therapeutic option [247,248].

8. Clinical Trials Against Flaviviruses

Flaviviruses represent a concern to global health since there are no specific antiviral drugs available for their treatment or prophylaxis. Promising preclinical candidates have been investigated for potential clinical benefits. Clinical trials employing FDA-approved drugs with potent antiviral activity in preclinical testing, such as balapiravir, chloroquine, lovastatin, and celgosivir, have failed to demonstrate efficacy in viral load reduction or beneficial clinical outcomes.
A plethora of antiviral compounds—both direct-acting and host-factor-targeting directed against distinct stages of the viral life cycle—are underway. As per the information retrieved on 1 July 2024 from https://clinicaltrials.gov, Table 7 lists the drugs targeting virion entrance and fusion, NS4B, NS5 polymerase, and host-directed antivirals. The antiviral efficacy of these compounds against JEV, ZIKV, YFV, and DENV is being assessed. Several of these compounds failed in the clinical trial, which thus called for the early conclusion of clinical trials, in addition to trials that have had to conclude early due to a lack of sufficient patients or funding.

9. Discussion and Conclusions

Flavivirus infections are a serious global health concern. Direct-acting antivirals with high effectiveness are desperately needed for both preventive and therapeutic action against illnesses caused by mosquito-borne flaviviruses. Different approaches have been implemented to develop prospective antiviral agents against flavivirus infection. These include (i) rational design derived from the crystal structures of viral proteins, (ii) screening the database of FDA-approved synthetic and natural compounds, (iii) testing of identified inhibitors effective against other human viruses, (iv) chemically altering the recognized viral inhibitors to enhance their therapeutic efficacy, (v) intravenous therapy with nucleic acids and immunoglobulins directed against viral genomes, (vi) antiviral peptides from combinatorial libraries by phage display, and (vii) exosomes (EVs) or microvesicles released from virus-infected cells. The implementation of these strategies resulted in the development of some successful antivirals, such as ZIKV-IG (20), ivermectin (251), Omr-IgG-am (NCT00068055), and UV-4B (167), assisting in the treatment of flavivirus infection. However, these approaches do have limitations. For instance, virus-directed inhibitors provide specificity, safety, and enhanced efficacy for treating flavivirus infections (NCT01856205), but this strategy often fails due to the emerging flaviviruses. In this scenario, host-directed inhibitors with potent antiviral activity (251,167) and a high barrier to resistance may be beneficial as they are poorly influenced by viral genetic polymorphisms. Nonetheless, these have caveats because they target host cellular functions that may be critical to cell survival and thus may be toxic. In addition, host-directed inhibitors are vulnerable to host genetic polymorphisms, which may hamper their ability to impair target protein activity.
The successful trial of IVIG provides clinical proof of concept for the development of antiviral treatments (NCT01856205). However, monoclonal antibodies with antiviral activity are also susceptible to mutations in the viral genome, which alters the pathogenic potential of the virus, resulting in the emergence of viral escape mutants, resistant to a specific monoclonal antibody (28). To combat this viral escape phenomenon, certain monoclonal antibodies targeting multiple viral epitopes may be used in conjugation to prevent the neutralization escape by the viruses. Similarly, engineering an RNA silencing mechanism aims to provide antiviral protection. The expression of amiRNAs could be employed to interfere with viral infections with high specificity (196–200). While many amiRNAs have demonstrated high antiviral efficiency, not all amiRNAs directed against viral targets efficiently restrict viral multiplication, leading to the emergence of a broad range of escape mutant viruses. Of note, peptide inhibitors are an appealing alternative as they are less expensive to create and safer than small-molecule- and antibody-based antiviral treatments (9,10,11,15). However, peptides have relatively short half-lives and immunogenicity.
Medicinal plants serve as an excellent source of biodiversity of novel natural compounds for drug discovery pipelines targeting viral infections (32–36). However, there are worries regarding absorption profiles, standardization, uniformity, and stability, in the early stages of drug discovery, to identify natural inhibitors with balanced pharmacokinetics (ADME—absorption, distribution, metabolism, and excretion). In a similar vein, exosomes represent an exciting intervention due to the absence of undesirable factors and their high biocompatibility with living tissues, such as primary human cell cultures and cell lines, as well as also being able to traverse biological barriers, such as the blood–brain barrier (BBB), which enhances bioavailability during neuroinflammation (206–210). Yet, the major considerations for clinically effective exosomes include scalability, repeatability, nontoxicity, property, and potency. Aside from that, the heterogeneity of exosomes, arising from their varied sources and cargo content, are hurdles for consistent therapeutic effectiveness and safety.
The challenges associated with these approaches warrant considerable exploration, necessitating an appropriate animal model showing clinical symptoms. Various animal models for flaviviruses have been tested. A perfect animal model must have reproducible viremia, be immuno-competent, and have viremia for all serotypes; unfortunately, no model meets all of these criteria. To date, mouse models such as AG129 [249] and IFNAR−/− [250] have been employed as a model of ZIKV pathogenesis to evaluate vaccines, therapeutics, and disease pathogenesis. The same mouse model as well as a tree shrew model [251] and A/J mice [252] have been described for DENV pathogenesis. Further, C57BL/6 mice [253], AG129 mice [254], and BALB/c mice [255] have been used as experimental models for JEV infection, whereas AG129 mice [256], IFNAR−/−, Syrian golden hamsters [257], and rhesus macaques [258] are primarily used as models for the yellow fever virus. Similarly, immunocompetent mice, hamsters, and geese are susceptible to WN virus infection [259,260,261].
Each of these experimental models resembles human disease to some extent. However, each has advantages and disadvantages when used in preclinical antiviral drug testing. Rodents are not natural flavivirus hosts; hence, they do not develop clinical signs like humans. Hamsters and non-human primates (NHPs) are natural hosts and serve as reservoirs for flaviviruses owing to physiological and genetic similarities with humans. Consequently, they have a benefit over rodents. Nevertheless, one drawback of NHP models is the availability of validated reagents for use with NHP materials.
A mere handful of inhibitors proven effective in animal models have advanced to clinical trials, and the findings cannot be easily extrapolated to humans. This is probably due to the paradoxical involvement of the host immune system in flavivirus infection. As a result, combination therapy trials incorporating both anti-inflammatory and antiviral drugs that have been evaluated in different animal models should be given due consideration.
Research must proceed rapidly, if appropriate, to in vivo investigations and clinical trials. The development of optimal anti-flaviviral drugs may benefit from the re-employment and repurposing of techniques and FDA-approved drugs. A thorough assessment of the drug combination is necessary to identify a beneficial synergy for heightened treatment. A promising method that may be applied to the treatment of flaviviruses is the use of multidrug combinations targeting different stages of viral replication and mitigating the impacts of antiviral resistance.
The insights gleaned from unsuccessful therapy trials may be useful in future research as novel compounds are clinically evaluated for their ability to counter flavivirus infection. The scientific methods for screening and studying inhibitors, which target the crucial host or viral elements involved in the flavivirus life cycle, along with the noteworthy advancements in molecular and structural virology, also offer opportunities and highlights for the advancement of future medical therapies for mosquito-borne flaviviruses. Countermeasures that limit the spread of flaviviruses and disease must be continuously developed and put into action. It is crucial for public health authorities to have surveillance systems that look into and manage the spatial localization of pathogens and their dissemination. This review seeks to highlight the significance of developing novel medications to treat these viruses, which are becoming more widespread worldwide. At present, the majority of nations with a flavivirus epidemic are found in tropical or subtropical regions without having recourse to hospitals or medical personnel. Nevertheless, this reality is about to change due to climate change [262], and even the wealthiest nations will face challenges due to a shortage of antiviral drugs.

Funding

This research received no external funding and the APC was waived off by MDPI publications.

Acknowledgments

We would like to acknowledge and thank the University of Texas Medical Branch, Galveston, Texas, for allowing us to use the subscribed software including Bio renders and Endnote for graphical enhancement of figures and referencing, respectively.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

25HC25-Hydroxylcholesterol
4-HPR4-Hydroxyphenyl retinamide
ACCAcetyl-coenzyme A carboxylase
ADEAntibody-dependent enhancement
AICAR5-Aminoimidazole-4-carboxamide ribonucleotide
AmiRNAsArtificial microRNAs
AMPKAdenosine monophosphate-activated protein kinase
CCapsid
CHIKVChikungunya
CMsConvoluted membranes
CQChloroquine
CSTCastanospermine
D2RDopamine D2 receptor
DEFA1Defensin alpha 1B
DENVDengue virus
DHODHDihydroorotate dehydrogenase
DNJDeoxynojirimycin
dsRNADouble-stranded RNA
EEnvelope
EDEE-dimer epitope
EDIIIDomain III of envelope proteins
EREndoplasmic reticulum
GTaseGuanylyltransferase
HCQHydroxychloroquine
HCV Hepatitis C virus
HmAbHuman monoclonal antibody
HTSHigh-throughput screening
IFITM3Interferon-induced transmembrane protein 3
JEVJapanese encephalitis virus
LDLRLow-density lipoprotein receptor
LGTVLangat virus
mAbMonoclonal antibodies
MBMethylene blue
MDDCsMonocyte-derived dendritic cells
MERS-CoVMiddle East Respiratory Syndrome coronavirus
MODVModoc virus
MTaseMethyltransferase
NDGANordihydroguaiaretic acid
NisNucleoside/nucleotide inhibitors
NITDN-sulfonylanthranilic acid derivative
NLSsNuclear localization sequences
NOAEL No observed adverse effect level
NPCNuclear pore complex
NSNon-structural
NSCsNeural stem cells
PCZProchlorperazine
PKPharmacokinetic
prM/MPrecursor membrane
RdRpRNA-dependent RNA polymerase
SAMS-adenosyl-L-methionine
SARStructure–activity relationship
shRNAShort hairpin RNA
siRNASmall interfering RNA
SLCSplit luciferase complementation
SREBPSterol regulatory element binding protein
TEMTransmission electron microscopic
TGNTrans-Golgi network
UTRUntranslated region
UV-4BUV-4 hydrochloride
VPsVesicular packets
VRCViral replication complex
WNVWest Nile virus
YFVYellow fever virus
ZIKVZika virus
ZIKV-IgHuman anti-ZIKV immunoglobulin

References

  1. Smit, J.M.; Moesker, B.; Rodenhuis-Zybert, I.; Wilschut, J. Flavivirus cell entry and membrane fusion. Viruses 2011, 3, 160–171. [Google Scholar] [CrossRef]
  2. Allison, S.L.; Stadler, K.; Mandl, C.W.; Kunz, C.; Heinz, F.X. Synthesis and secretion of recombinant tick-borne encephalitis virus protein E in soluble and particulate form. J. Virol. 1995, 69, 5816–5820. [Google Scholar] [CrossRef] [PubMed]
  3. Lindenbach, B.D.; Rice, C.M. Molecular biology of flaviviruses. Adv. Virus Res. 2003, 59, 23–61. [Google Scholar] [CrossRef]
  4. Brinton, M.A. The molecular biology of West Nile Virus: A new invader of the western hemisphere. Annu. Rev. Microbiol. 2002, 56, 371–402. [Google Scholar] [CrossRef] [PubMed]
  5. Yu, Y.; Deng, Y.Q.; Zou, P.; Wang, Q.; Dai, Y.; Yu, F.; Du, L.; Zhang, N.N.; Tian, M.; Hao, J.N.; et al. A peptide-based viral inactivator inhibits Zika virus infection in pregnant mice and fetuses. Nat. Commun. 2017, 8, 15672. [Google Scholar] [CrossRef] [PubMed]
  6. Qian, X.; Wu, B.; Tang, H.; Luo, Z.; Xu, Z.; Ouyang, S.; Li, X.; Xie, J.; Yi, Z.; Leng, Q.; et al. Rifapentine is an entry and replication inhibitor against yellow fever virus both in vitro and in vivo. Emerg. Microbes Infect. 2022, 11, 873–884. [Google Scholar] [CrossRef] [PubMed]
  7. Qian, X.; Qi, Z. Mosquito-Borne Flaviviruses and Current Therapeutic Advances. Viruses 2022, 14, 1226. [Google Scholar] [CrossRef]
  8. Mediaty, A.; Neuber, K. Total and specific serum IgE decreases with age in patients with allergic rhinitis, asthma and insect allergy but not in patients with atopic dermatitis. Immun. Ageing 2005, 2, 9. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, L.; Liu, Y.; Wang, S.; Sun, J.; Wang, P.; Xin, Q.; Zhang, L.; Xiao, G.; Wang, W. Antiviral activity of peptide inhibitors derived from the protein E stem against Japanese encephalitis and Zika viruses. Antivir. Res. 2017, 141, 140–149. [Google Scholar] [CrossRef] [PubMed]
  10. Alhoot, M.A.; Rathinam, A.K.; Wang, S.M.; Manikam, R.; Sekaran, S.D. Inhibition of dengue virus entry into target cells using synthetic antiviral peptides. Int. J. Med. Sci. 2013, 10, 719–729. [Google Scholar] [CrossRef] [PubMed]
  11. Isa, D.M.; Chin, S.P.; Chong, W.L.; Zain, S.M.; Rahman, N.A.; Lee, V.S. Dynamics and binding interactions of peptide inhibitors of dengue virus entry. J. Biol. Phys. 2019, 45, 63–76. [Google Scholar] [CrossRef] [PubMed]
  12. Panya, A.; Bangphoomi, K.; Choowongkomon, K.; Yenchitsomanus, P.T. Peptide inhibitors against dengue virus infection. Chem. Biol. Drug Des. 2014, 84, 148–157. [Google Scholar] [CrossRef] [PubMed]
  13. Li, P.C.; Jang, J.; Hsia, C.Y.; Groomes, P.V.; Lian, W.; de Wispelaere, M.; Pitts, J.D.; Wang, J.; Kwiatkowski, N.; Gray, N.S.; et al. Small Molecules Targeting the Flavivirus E Protein with Broad-Spectrum Activity and Antiviral Efficacy in Vivo. ACS Infect. Dis. 2019, 5, 460–472. [Google Scholar] [CrossRef] [PubMed]
  14. Robinson, L.N.; Tharakaraman, K.; Rowley, K.J.; Costa, V.V.; Chan, K.R.; Wong, Y.H.; Ong, L.C.; Tan, H.C.; Koch, T.; Cain, D.; et al. Structure-Guided Design of an Anti-dengue Antibody Directed to a Non-immunodominant Epitope. Cell 2015, 162, 493–504. [Google Scholar] [CrossRef]
  15. Fibriansah, G.; Ibarra, K.D.; Ng, T.S.; Smith, S.A.; Tan, J.L.; Lim, X.N.; Ooi, J.S.; Kostyuchenko, V.A.; Wang, J.; de Silva, A.M.; et al. DENGUE VIRUS. Cryo-EM structure of an antibody that neutralizes dengue virus type 2 by locking E protein dimers. Science 2015, 349, 88–91. [Google Scholar] [CrossRef]
  16. Komarasamy, T.V.; Adnan, N.A.A.; James, W.; Balasubramaniam, V.R. Finding a chink in the armor: Update, limitations, and challenges toward successful antivirals against flaviviruses. PLoS Negl. Trop. Dis. 2022, 16, e0010291. [Google Scholar] [CrossRef] [PubMed]
  17. White, J.; Tunga, P.; Anderson, D.M.; Iledan, K.; Loreth, T.; Parrera, G.S.; Astacio, H.; Drobic, B.; Richardson, J.S. Results of a Double-Blind, Randomized, Placebo-Controlled Phase 1 Study to Evaluate the Safety and Pharmacokinetics of Anti-Zika Virus Immunoglobulin. Am. J. Trop. Med. Hyg. 2021, 105, 1552–1562. [Google Scholar] [CrossRef]
  18. Hrobowski, Y.M.; Garry, R.F.; Michael, S.F. Peptide inhibitors of dengue virus and West Nile virus infectivity. Virol. J. 2005, 2, 49. [Google Scholar] [CrossRef] [PubMed]
  19. Beigel, J.H.; Nordstrom, J.L.; Pillemer, S.R.; Roncal, C.; Goldwater, D.R.; Li, H.; Holland, P.C.; Johnson, S.; Stein, K.; Koenig, S. Safety and pharmacokinetics of single intravenous dose of MGAWN1, a novel monoclonal antibody to West Nile virus. Antimicrob. Agents Chemother. 2010, 54, 2431–2436. [Google Scholar] [CrossRef]
  20. de Wispelaere, M.; Lian, W.; Potisopon, S.; Li, P.C.; Jang, J.; Ficarro, S.B.; Clark, M.J.; Zhu, X.; Kaplan, J.B.; Pitts, J.D.; et al. Inhibition of Flaviviruses by Targeting a Conserved Pocket on the Viral Envelope Protein. Cell Chem. Biol. 2018, 25, 1006–1016.e8. [Google Scholar] [CrossRef]
  21. Wang, J.; Bardelli, M.; Espinosa, D.A.; Pedotti, M.; Ng, T.S.; Bianchi, S.; Simonelli, L.; Lim, E.X.Y.; Foglierini, M.; Zatta, F.; et al. A Human Bi-specific Antibody against Zika Virus with High Therapeutic Potential. Cell 2017, 171, e215. [Google Scholar] [CrossRef]
  22. Sapparapu, G.; Fernandez, E.; Kose, N.; Bin, C.; Fox, J.M.; Bombardi, R.G.; Zhao, H.; Nelson, C.A.; Bryan, A.L.; Barnes, T.; et al. Neutralizing human antibodies prevent Zika virus replication and fetal disease in mice. Nature 2016, 540, 443–447. [Google Scholar] [CrossRef]
  23. Barba-Spaeth, G.; Dejnirattisai, W.; Rouvinski, A.; Vaney, M.C.; Medits, I.; Sharma, A.; Simon-Loriere, E.; Sakuntabhai, A.; Cao-Lormeau, V.M.; Haouz, A.; et al. Structural basis of potent Zika-dengue virus antibody cross-neutralization. Nature 2016, 536, 48–53. [Google Scholar] [CrossRef]
  24. Goo, L.; Debbink, K.; Kose, N.; Sapparapu, G.; Doyle, M.P.; Wessel, A.W.; Richner, J.M.; Burgomaster, K.E.; Larman, B.C.; Dowd, K.A.; et al. A protective human monoclonal antibody targeting the West Nile virus E protein preferentially recognizes mature virions. Nat. Microbiol. 2019, 4, 71–77. [Google Scholar] [CrossRef]
  25. Gao, Y.; Tai, W.; Wang, N.; Li, X.; Jiang, S.; Debnath, A.K.; Du, L.; Chen, S. Identification of Novel Natural Products as Effective and Broad-Spectrum Anti-Zika Virus Inhibitors. Viruses 2019, 11, 1019. [Google Scholar] [CrossRef] [PubMed]
  26. Mounce, B.C.; Cesaro, T.; Carrau, L.; Vallet, T.; Vignuzzi, M. Curcumin inhibits Zika and chikungunya virus infection by inhibiting cell binding. Antivir. Res. 2017, 142, 148–157. [Google Scholar] [CrossRef]
  27. Padilla, S.L.; Rodriguez, A.; Gonzales, M.M.; Gallego, G.J.; Castano, O.J. Inhibitory effects of curcumin on dengue virus type 2-infected cells in vitro. Arch. Virol. 2014, 159, 573–579. [Google Scholar] [CrossRef]
  28. Yang, C.C.; Hu, H.S.; Lin, H.M.; Wu, P.S.; Wu, R.H.; Tian, J.N.; Wu, S.H.; Tsou, L.K.; Song, J.S.; Chen, H.W.; et al. A novel flavivirus entry inhibitor, BP34610, discovered through high-throughput screening with dengue reporter viruses. Antivir. Res. 2019, 172, 104636. [Google Scholar] [CrossRef]
  29. Abdul Ahmad, S.A.; Palanisamy, U.D.; Tejo, B.A.; Chew, M.F.; Tham, H.W.; Syed Hassan, S. Geraniin extracted from the rind of Nephelium lappaceum binds to dengue virus type-2 envelope protein and inhibits early stage of virus replication. Virol. J. 2017, 14, 229. [Google Scholar] [CrossRef]
  30. Abdul Ahmad, S.A.; Palanisamy, U.D.; Khoo, J.J.; Dhanoa, A.; Syed Hassan, S. Efficacy of geraniin on dengue virus type-2 infected BALB/c mice. Virol. J. 2019, 16, 26. [Google Scholar] [CrossRef] [PubMed]
  31. Jia, F.; Zou, G.; Fan, J.; Yuan, Z. Identification of palmatine as an inhibitor of West Nile virus. Arch. Virol. 2010, 155, 1325–1329. [Google Scholar] [CrossRef] [PubMed]
  32. Simanjuntak, Y.; Liang, J.J.; Lee, Y.L.; Lin, Y.L. Repurposing of prochlorperazine for use against dengue virus infection. J. Infect. Dis. 2015, 211, 394–404. [Google Scholar] [CrossRef]
  33. Barrows, N.J.; Campos, R.K.; Powell, S.T.; Prasanth, K.R.; Schott-Lerner, G.; Soto-Acosta, R.; Galarza-Munoz, G.; McGrath, E.L.; Urrabaz-Garza, R.; Gao, J.; et al. A Screen of FDA-Approved Drugs for Inhibitors of Zika Virus Infection. Cell Host Microbe 2016, 20, 259–270. [Google Scholar] [CrossRef]
  34. Rausch, K.; Hackett, B.A.; Weinbren, N.L.; Reeder, S.M.; Sadovsky, Y.; Hunter, C.A.; Schultz, D.C.; Coyne, C.B.; Cherry, S. Screening Bioactives Reveals Nanchangmycin as a Broad Spectrum Antiviral Active against Zika Virus. Cell Rep. 2017, 18, 804–815. [Google Scholar] [CrossRef] [PubMed]
  35. Abrams, R.P.M.; Yasgar, A.; Teramoto, T.; Lee, M.H.; Dorjsuren, D.; Eastman, R.T.; Malik, N.; Zakharov, A.V.; Li, W.; Bachani, M.; et al. Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors. Proc. Natl. Acad. Sci. USA 2020, 117, 31365–31375. [Google Scholar] [CrossRef]
  36. Pu, S.Y.; Xiao, F.; Schor, S.; Bekerman, E.; Zanini, F.; Barouch-Bentov, R.; Nagamine, C.M.; Einav, S. Feasibility and biological rationale of repurposing sunitinib and erlotinib for dengue treatment. Antivir. Res. 2018, 155, 67–75. [Google Scholar] [CrossRef]
  37. Li, C.; Deng, Y.Q.; Wang, S.; Ma, F.; Aliyari, R.; Huang, X.Y.; Zhang, N.N.; Watanabe, M.; Dong, H.L.; Liu, P.; et al. 25-Hydroxycholesterol Protects Host against Zika Virus Infection and Its Associated Microcephaly in a Mouse Model. Immunity 2017, 46, 446–456. [Google Scholar] [CrossRef]
  38. Savarino, A.; Boelaert, J.R.; Cassone, A.; Majori, G.; Cauda, R. Effects of chloroquine on viral infections: An old drug against today’s diseases? Lancet Infect. Dis. 2003, 3, 722–727. [Google Scholar] [CrossRef]
  39. Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef] [PubMed]
  40. Tricou, V.; Minh, N.N.; Van, T.P.; Lee, S.J.; Farrar, J.; Wills, B.; Tran, H.T.; Simmons, C.P. A randomized controlled trial of chloroquine for the treatment of dengue in Vietnamese adults. PLoS Negl. Trop. Dis. 2010, 4, e785. [Google Scholar] [CrossRef]
  41. Dyall, J.; Gross, R.; Kindrachuk, J.; Johnson, R.F.; Olinger, G.G.J.; Hensley, L.E.; Frieman, M.B.; Jahrling, P.B. Middle East Respiratory Syndrome and Severe Acute Respiratory Syndrome: Current Therapeutic Options and Potential Targets for Novel Therapies. Drugs 2017, 77, 1935–1966. [Google Scholar] [CrossRef]
  42. Michael, F.S.; Isern, S. Flavivirus-Binding, Entry-Obstructing, Protease-Resistant Peptide (RI57). U.S. Patent 10639380 B2, 5 May 2020. [Google Scholar]
  43. Bailey, M.J.; Broecker, F.; Freyn, A.W.; Choi, A.; Brown, J.A.; Fedorova, N.; Simon, V.; Lim, J.K.; Evans, M.J.; Garcia-Sastre, A.; et al. Human Monoclonal Antibodies Potently Neutralize Zika Virus and Select for Escape Mutations on the Lateral Ridge of the Envelope Protein. J. Virol. 2019, 93, e00405-19. [Google Scholar] [CrossRef]
  44. Saxena, P.R.; Lawang, A. A comparison of cardiovascular and smooth muscle effects of 5-hydroxytryptamine and 5-carboxamidotryptamine, a selective agonist of 5-HT1 receptors. Arch. Int. Pharmacodyn. Ther. 1985, 277, 235–252. [Google Scholar]
  45. Badani, H.; Garry, R.F.; Wimley, W.C. Peptide entry inhibitors of enveloped viruses: The importance of interfacial hydrophobicity. Biochim. Biophys. Acta. 2014, 1838, 2180–2197. [Google Scholar] [CrossRef]
  46. Shoji-Kawata, S.; Sumpter, R.; Leveno, M.; Campbell, G.R.; Zou, Z.; Kinch, L.; Wilkins, A.D.; Sun, Q.; Pallauf, K.; MacDuff, D.; et al. Identification of a candidate therapeutic autophagy-inducing peptide. Nature 2013, 494, 201–206. [Google Scholar] [CrossRef] [PubMed]
  47. Focosi, D.; McConnell, S.; Casadevall, A.; Cappello, E.; Valdiserra, G.; Tuccori, M. Monoclonal antibody therapies against SARS-CoV-2. Lancet Infect. Dis. 2022, 22, e311–e326. [Google Scholar] [CrossRef] [PubMed]
  48. Jiang, S.; Du, L. Advances in the research and development of therapeutic antibodies against the Zika virus. Cell. Mol. Immunol. 2019, 16, 96–97. [Google Scholar] [CrossRef]
  49. Mancini, N. Monoclonal Antibodies and Flaviviruses: A Possible Option? mBio 2022, 13, e0082422. [Google Scholar] [CrossRef] [PubMed]
  50. Thomas, S.; Smatti, M.K.; Ouhtit, A.; Cyprian, F.S.; Almaslamani, M.A.; Thani, A.A.; Yassine, H.M. Antibody-Dependent Enhancement (ADE) and the role of complement system in disease pathogenesis. Mol. Immunol. 2022, 152, 172–182. [Google Scholar] [CrossRef] [PubMed]
  51. Roy, A.; Lim, L.; Srivastava, S.; Lu, Y.; Song, J. Solution conformations of Zika NS2B-NS3pro and its inhibition by natural products from edible plants. PLoS ONE 2017, 12, e0180632. [Google Scholar] [CrossRef] [PubMed]
  52. Allard, P.M.; Dau, E.T.; Eydoux, C.; Guillemot, J.C.; Dumontet, V.; Poullain, C.; Canard, B.; Gueritte, F.; Litaudon, M. Alkylated flavanones from the bark of Cryptocarya chartacea as dengue virus NS5 polymerase inhibitors. J. Nat. Prod. 2011, 74, 2446–2453. [Google Scholar] [CrossRef]
  53. Nawa, M.; Takasaki, T.; Yamada, K.I.; Kurane, I.; Akatsuka, T. Interference in Japanese encephalitis virus infection of Vero cells by a cationic amphiphilic drug, chlorpromazine. J. Gen. Virol. 2003, 84, 1737–1741. [Google Scholar] [CrossRef] [PubMed]
  54. Jimenez de Oya, N.; Blazquez, A.B.; Casas, J.; Saiz, J.C.; Martin-Acebes, M.A. Direct Activation of Adenosine Monophosphate-Activated Protein Kinase (AMPK) by PF-06409577 Inhibits Flavivirus Infection through Modification of Host Cell Lipid Metabolism. Antimicrob. Agents Chemother. 2018, 62, e00360-18. [Google Scholar] [CrossRef] [PubMed]
  55. Jimenez de Oya, N.; Esler, W.P.; Huard, K.; El-Kattan, A.F.; Karamanlidis, G.; Blazquez, A.B.; Ramos-Ibeas, P.; Escribano-Romero, E.; Louloudes-Lazaro, A.; Casas, J.; et al. Targeting host metabolism by inhibition of acetyl-Coenzyme A carboxylase reduces flavivirus infection in mouse models. Emerg. Microbes Infect. 2019, 8, 624–636. [Google Scholar] [CrossRef]
  56. Huang, Y.; Li, Y.; Zhang, H.; Zhao, R.; Jing, R.; Xu, Y.; He, M.; Peer, J.; Kim, Y.C.; Luo, J.; et al. Zika virus propagation and release in human fetal astrocytes can be suppressed by neutral sphingomyelinase-2 inhibitor GW4869. Cell Discov. 2018, 4, 19. [Google Scholar] [CrossRef] [PubMed]
  57. Martin-Acebes, M.A.; Merino-Ramos, T.; Blazquez, A.B.; Casas, J.; Escribano-Romero, E.; Sobrino, F.; Saiz, J.C. The composition of West Nile virus lipid envelope unveils a role of sphingolipid metabolism in flavivirus biogenesis. J. Virol. 2014, 88, 12041–12054. [Google Scholar] [CrossRef]
  58. Navarro-Sanchez, E.; Altmeyer, R.; Amara, A.; Schwartz, O.; Fieschi, F.; Virelizier, J.L.; Arenzana-Seisdedos, F.; Despres, P. Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Rep. 2003, 4, 723–728. [Google Scholar] [CrossRef] [PubMed]
  59. Xu, M.; Lee, E.M.; Wen, Z.; Cheng, Y.; Huang, W.K.; Qian, X.; Tcw, J.; Kouznetsova, J.; Ogden, S.C.; Hammack, C.; et al. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat. Med. 2016, 22, 1101–1107. [Google Scholar] [CrossRef]
  60. Yang, C.F.; Gopula, B.; Liang, J.J.; Li, J.K.; Chen, S.Y.; Lee, Y.L.; Chen, C.S.; Lin, Y.L. Novel AR-12 derivatives, P12-23 and P12-34, inhibit flavivirus replication by blocking host de novo pyrimidine biosynthesis. Emerg. Microbes Infect. 2018, 7, 187. [Google Scholar] [CrossRef] [PubMed]
  61. Chu, J.J.; Ng, M.L. Infectious entry of West Nile virus occurs through a clathrin-mediated endocytic pathway. J. Virol. 2004, 78, 10543–10555. [Google Scholar] [CrossRef]
  62. Martin-Acebes, M.A.; Jimenez de Oya, N.; Saiz, J.C. Lipid Metabolism as a Source of Druggable Targets for Antiviral Discovery against Zika and Other Flaviviruses. Pharmaceuticals 2019, 12, 97. [Google Scholar] [CrossRef]
  63. Farfan-Morales, C.N.; Cordero-Rivera, C.D.; Osuna-Ramos, J.F.; Monroy-Munoz, I.E.; De Jesus-Gonzalez, L.A.; Munoz-Medina, J.E.; Hurtado-Monzon, A.M.; Reyes-Ruiz, J.M.; Del Angel, R.M. The antiviral effect of metformin on zika and dengue virus infection. Sci. Rep. 2021, 11, 8743. [Google Scholar] [CrossRef] [PubMed]
  64. Htun, H.L.; Yeo, T.W.; Tam, C.C.; Pang, J.; Leo, Y.S.; Lye, D.C. Metformin Use and Severe Dengue in Diabetic Adults. Sci. Rep. 2018, 8, 3344. [Google Scholar] [CrossRef]
  65. Li, Z.; Brecher, M.; Deng, Y.Q.; Zhang, J.; Sakamuru, S.; Liu, B.; Huang, R.; Koetzner, C.A.; Allen, C.A.; Jones, S.A.; et al. Existing drugs as broad-spectrum and potent inhibitors for Zika virus by targeting NS2B-NS3 interaction. Cell Res. 2017, 27, 1046–1064. [Google Scholar] [CrossRef] [PubMed]
  66. Akey, D.L.; Brown, W.C.; Jose, J.; Kuhn, R.J.; Smith, J.L. Structure-guided insights on the role of NS1 in flavivirus infection. Bioessays 2015, 37, 489–494. [Google Scholar] [CrossRef] [PubMed]
  67. Welsch, S.; Miller, S.; Romero-Brey, I.; Merz, A.; Bleck, C.K.; Walther, P.; Fuller, S.D.; Antony, C.; Krijnse-Locker, J.; Bartenschlager, R. Composition and three-dimensional architecture of the dengue virus replication and assembly sites. Cell Host Microbe 2009, 5, 365–375. [Google Scholar] [CrossRef]
  68. Songprakhon, P.; Thaingtamtanha, T.; Limjindaporn, T.; Puttikhunt, C.; Srisawat, C.; Luangaram, P.; Dechtawewat, T.; Uthaipibull, C.; Thongsima, S.; Yenchitsomanus, P.T.; et al. Peptides targeting dengue viral nonstructural protein 1 inhibit dengue virus production. Sci. Rep. 2020, 10, 12933. [Google Scholar] [CrossRef] [PubMed]
  69. Sun, E.C.; Ma, J.N.; Liu, N.H.; Yang, T.; Zhao, J.; Geng, H.W.; Wang, L.F.; Qin, Y.L.; Bu, Z.G.; Yang, Y.H.; et al. Identification of two linear B-cell epitopes from West Nile virus NS1 by screening a phage-displayed random peptide library. BMC Microbiol. 2011, 11, 160. [Google Scholar] [CrossRef]
  70. Raza, S.; Abbas, G.; Azam, S.S. Screening Pipeline for Flavivirus Based Inhibitors for Zika Virus NS1. IEEE/ACM Trans. Comput. Biol. Bioinform. 2020, 17, 1751–1761. [Google Scholar] [CrossRef]
  71. Bailey, M.J.; Duehr, J.; Dulin, H.; Broecker, F.; Brown, J.A.; Arumemi, F.O.; Bermudez Gonzalez, M.C.; Leyva-Grado, V.H.; Evans, M.J.; Simon, V.; et al. Human antibodies targeting Zika virus NS1 provide protection against disease in a mouse model. Nat. Commun. 2018, 9, 4560. [Google Scholar] [CrossRef]
  72. Biering, S.B.; Akey, D.L.; Wong, M.P.; Brown, W.C.; Lo, N.T.N.; Puerta-Guardo, H.; Tramontini Gomes de Sousa, F.; Wang, C.; Konwerski, J.R.; Espinosa, D.A.; et al. Structural basis for antibody inhibition of flavivirus NS1-triggered endothelial dysfunction. Science 2021, 371, 194–200. [Google Scholar] [CrossRef] [PubMed]
  73. Courageot, M.P.; Frenkiel, M.P.; Dos Santos, C.D.; Deubel, V.; Despres, P. Alpha-glucosidase inhibitors reduce dengue virus production by affecting the initial steps of virion morphogenesis in the endoplasmic reticulum. J. Virol. 2000, 74, 564–572. [Google Scholar] [CrossRef]
  74. Wu, S.F.; Lee, C.J.; Liao, C.L.; Dwek, R.A.; Zitzmann, N.; Lin, Y.L. Antiviral effects of an iminosugar derivative on flavivirus infections. J. Virol. 2002, 76, 3596–3604. [Google Scholar] [CrossRef] [PubMed]
  75. Edeling, M.A.; Diamond, M.S.; Fremont, D.H. Structural basis of Flavivirus NS1 assembly and antibody recognition. Proc. Natl. Acad. Sci. USA 2014, 111, 4285–4290. [Google Scholar] [CrossRef]
  76. Halstead, S.B. Stumbles on the path to dengue control. Lancet Infect. Dis. 2014, 14, 661–662. [Google Scholar] [CrossRef] [PubMed]
  77. Montes-Grajales, D.; Puerta-Guardo, H.; Espinosa, D.A.; Harris, E.; Caicedo-Torres, W.; Olivero-Verbel, J.; Martinez-Romero, E. In silico drug repurposing for the identification of potential candidate molecules against arboviruses infection. Antivir. Res. 2020, 173, 104668. [Google Scholar] [CrossRef]
  78. Yuan, S.; Chan, J.F.; den-Haan, H.; Chik, K.K.; Zhang, A.J.; Chan, C.C.; Poon, V.K.; Yip, C.C.; Mak, W.W.; Zhu, Z.; et al. Structure-based discovery of clinically approved drugs as Zika virus NS2B-NS3 protease inhibitors that potently inhibit Zika virus infection in vitro and in vivo. Antivir. Res. 2017, 145, 33–43. [Google Scholar] [CrossRef] [PubMed]
  79. Li, Z.; Xu, J.; Lang, Y.; Fan, X.; Kuo, L.; D’Brant, L.; Hu, S.; Samrat, S.K.; Trudeau, N.; Tharappel, A.M.; et al. JMX0207, a Niclosamide Derivative with Improved Pharmacokinetics, Suppresses Zika Virus Infection Both In Vitro and In Vivo. ACS Infect. Dis. 2020, 6, 2616–2628. [Google Scholar] [CrossRef]
  80. Bhakat, S.; Delang, L.; Kaptein, S.; Neyts, J.; Leyssen, P.; Jayaprakash, V. Reaching beyond HIV/HCV: Nelfinavir as a potential starting point for broad-spectrum protease inhibitors against dengue and chikungunya virus. RSC Adv. 2015, 5, 85938–85949. [Google Scholar] [CrossRef]
  81. Rassias, G.; Zogali, V.; Swarbrick, C.M.D.; Ki Chan, K.W.; Chan, S.A.; Gwee, C.P.; Wang, S.; Kaplanai, E.; Canko, A.; Kiousis, D.; et al. Cell-active carbazole derivatives as inhibitors of the zika virus protease. Eur. J. Med. Chem. 2019, 180, 536–545. [Google Scholar] [CrossRef] [PubMed]
  82. Cabarcas-Montalvo, M.; Maldonado-Rojas, W.; Montes-Grajales, D.; Bertel-Sevilla, A.; Wagner-Dobler, I.; Sztajer, H.; Reck, M.; Flechas-Alarcon, M.; Ocazionez, R.; Olivero-Verbel, J. Discovery of antiviral molecules for dengue: In silico search and biological evaluation. Eur. J. Med. Chem. 2016, 110, 87–97. [Google Scholar] [CrossRef] [PubMed]
  83. Brecher, M.; Li, Z.; Liu, B.; Zhang, J.; Koetzner, C.A.; Alifarag, A.; Jones, S.A.; Lin, Q.; Kramer, L.D.; Li, H. A conformational switch high-throughput screening assay and allosteric inhibition of the flavivirus NS2B-NS3 protease. PLoS Pathog. 2017, 13, e1006411. [Google Scholar] [CrossRef] [PubMed]
  84. Cui, X.; Zhou, R.; Huang, C.; Zhang, R.; Wang, J.; Zhang, Y.; Ding, J.; Li, X.; Zhou, J.; Cen, S. Identification of Theaflavin-3,3’-Digallate as a Novel Zika Virus Protease Inhibitor. Front. Pharmacol. 2020, 11, 514313. [Google Scholar] [CrossRef] [PubMed]
  85. Raut, R.; Beesetti, H.; Tyagi, P.; Khanna, I.; Jain, S.K.; Jeankumar, V.U.; Yogeeswari, P.; Sriram, D.; Swaminathan, S. A small molecule inhibitor of dengue virus type 2 protease inhibits the replication of all four dengue virus serotypes in cell culture. Virol. J. 2015, 12, 16. [Google Scholar] [CrossRef] [PubMed]
  86. Aleshin, A.E.; Shiryaev, S.A.; Strongin, A.Y.; Liddington, R.C. Structural evidence for regulation and specificity of flaviviral proteases and evolution of the Flaviviridae fold. Protein Sci. 2007, 16, 795–806. [Google Scholar] [CrossRef] [PubMed]
  87. Kumar, A.; Liang, B.; Aarthy, M.; Singh, S.K.; Garg, N.; Mysorekar, I.U.; Giri, R. Hydroxychloroquine Inhibits Zika Virus NS2B-NS3 Protease. ACS Omega 2018, 3, 18132–18141. [Google Scholar] [CrossRef] [PubMed]
  88. Yang, Y.P.; Hu, L.F.; Zheng, H.F.; Mao, C.J.; Hu, W.D.; Xiong, K.P.; Wang, F.; Liu, C.F. Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol. Sin. 2013, 34, 625–635. [Google Scholar] [CrossRef]
  89. Li, Z.; Lang, Y.; Sakamuru, S.; Samrat, S.; Trudeau, N.; Kuo, L.; Rugenstein, N.; Tharappel, A.; D’Brant, L.; Koetzner, C.A.; et al. Methylene blue is a potent and broad-spectrum inhibitor against Zika virus in vitro and in vivo. Emerg. Microbes Infect. 2020, 9, 2404–2416. [Google Scholar] [CrossRef] [PubMed]
  90. Li, Z.; Xu, J.; Lang, Y.; Wu, X.; Hu, S.; Samrat, S.K.; Tharappel, A.M.; Kuo, L.; Butler, D.; Song, Y.; et al. In vitro and in vivo characterization of erythrosin B and derivatives against Zika virus. Acta Pharm. Sin. B 2022, 12, 1662–1670. [Google Scholar] [CrossRef]
  91. Li, Z.; Sakamuru, S.; Huang, R.; Brecher, M.; Koetzner, C.A.; Zhang, J.; Chen, H.; Qin, C.F.; Zhang, Q.Y.; Zhou, J.; et al. Erythrosin B is a potent and broad-spectrum orthosteric inhibitor of the flavivirus NS2B-NS3 protease. Antivir. Res. 2018, 150, 217–225. [Google Scholar] [CrossRef] [PubMed]
  92. Mastrangelo, E.; Pezzullo, M.; De Burghgraeve, T.; Kaptein, S.; Pastorino, B.; Dallmeier, K.; de Lamballerie, X.; Neyts, J.; Hanson, A.M.; Frick, D.N.; et al. Ivermectin is a potent inhibitor of flavivirus replication specifically targeting NS3 helicase activity: New prospects for an old drug. J. Antimicrob. Chemother. 2012, 67, 1884–1894. [Google Scholar] [CrossRef] [PubMed]
  93. Ketkar, H.; Yang, L.; Wormser, G.P.; Wang, P. Lack of efficacy of ivermectin for prevention of a lethal Zika virus infection in a murine system. Diagn. Microbiol. Infect. Dis. 2019, 95, 38–40. [Google Scholar] [CrossRef] [PubMed]
  94. Caly, L.; Druce, J.D.; Catton, M.G.; Jans, D.A.; Wagstaff, K.M. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antivir. Res 2020, 178, 104787. [Google Scholar] [CrossRef]
  95. Ci, Y.; Yao, B.; Yue, K.; Yang, Y.; Xu, C.; Li, D.F.; Qin, C.F.; Shi, L. Bortezomib inhibits ZIKV/DENV by interfering with viral polyprotein cleavage via the ERAD pathway. Cell Chem. Biol. 2023, 30, 527–539.e5. [Google Scholar] [CrossRef] [PubMed]
  96. Pathak, N.; Lai, M.L.; Chen, W.Y.; Hsieh, B.W.; Yu, G.Y.; Yang, J.M. Pharmacophore anchor models of flaviviral NS3 proteases lead to drug repurposing for DENV infection. BMC Bioinform. 2017, 18, 548. [Google Scholar] [CrossRef]
  97. Byrd, C.M.; Grosenbach, D.W.; Berhanu, A.; Dai, D.; Jones, K.F.; Cardwell, K.B.; Schneider, C.; Yang, G.; Tyavanagimatt, S.; Harver, C.; et al. Novel benzoxazole inhibitor of dengue virus replication that targets the NS3 helicase. Antimicrob. Agents Chemother. 2013, 57, 1902–1912. [Google Scholar] [CrossRef]
  98. Yao, Y.; Huo, T.; Lin, Y.L.; Nie, S.; Wu, F.; Hua, Y.; Wu, J.; Kneubehl, A.R.; Vogt, M.B.; Rico-Hesse, R.; et al. Discovery, X-ray Crystallography and Antiviral Activity of Allosteric Inhibitors of Flavivirus NS2B-NS3 Protease. J. Am. Chem. Soc. 2019, 141, 6832–6836. [Google Scholar] [CrossRef]
  99. Sweeney, N.L.; Hanson, A.M.; Mukherjee, S.; Ndjomou, J.; Geiss, B.J.; Steel, J.J.; Frankowski, K.J.; Li, K.; Schoenen, F.J.; Frick, D.N. Benzothiazole and Pyrrolone Flavivirus Inhibitors Targeting the Viral Helicase. ACS Infect. Dis. 2015, 1, 140–148. [Google Scholar] [CrossRef]
  100. Basavannacharya, C.; Vasudevan, S.G. Suramin inhibits helicase activity of NS3 protein of dengue virus in a fluorescence-based high throughput assay format. Biochem. Biophys. Res. Commun. 2014, 453, 539–544. [Google Scholar] [CrossRef] [PubMed]
  101. Shiryaev, S.A.; Farhy, C.; Pinto, A.; Huang, C.T.; Simonetti, N.; Elong Ngono, A.; Dewing, A.; Shresta, S.; Pinkerton, A.B.; Cieplak, P.; et al. Characterization of the Zika virus two-component NS2B-NS3 protease and structure-assisted identification of allosteric small-molecule antagonists. Antivir. Res. 2017, 143, 218–229. [Google Scholar] [CrossRef] [PubMed]
  102. Nobori, H.; Toba, S.; Yoshida, R.; Hall, W.W.; Orba, Y.; Sawa, H.; Sato, A. Identification of Compound-B, a novel anti-dengue virus agent targeting the non-structural protein 4A. Antivir. Res. 2018, 155, 60–66. [Google Scholar] [CrossRef] [PubMed]
  103. Kaptein, S.J.F.; Goethals, O.; Kiemel, D.; Marchand, A.; Kesteleyn, B.; Bonfanti, J.F.; Bardiot, D.; Stoops, B.; Jonckers, T.H.M.; Dallmeier, K.; et al. Publisher Correction: A pan-serotype dengue virus inhibitor targeting the NS3-NS4B interaction. Nature 2021, 599, E2. [Google Scholar] [CrossRef] [PubMed]
  104. Goethals, O.; Voge, N.V.; Kesteleyn, B.; Chaltin, P.; Jinks, T.; De Marez, T.; Koul, A.; Draghia-Akli, R.; Neyts, J.; Van Loock, M. A pan-serotype antiviral to prevent and treat dengue: A journey from discovery to clinical development driven by public-private partnerships. Antivir. Res. 2023, 210, 105495. [Google Scholar] [CrossRef] [PubMed]
  105. Moquin, S.A.; Simon, O.; Karuna, R.; Lakshminarayana, S.B.; Yokokawa, F.; Wang, F.; Saravanan, C.; Zhang, J.; Day, C.W.; Chan, K.; et al. NITD-688, a pan-serotype inhibitor of the dengue virus NS4B protein, shows favorable pharmacokinetics and efficacy in preclinical animal models. Sci. Transl. Med. 2021, 13, eabb2181. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, S.; Liu, Y.; Guo, J.; Wang, P.; Zhang, L.; Xiao, G.; Wang, W. Screening of FDA-Approved Drugs for Inhibitors of Japanese Encephalitis Virus Infection. J. Virol. 2017, 91, e01055-17. [Google Scholar] [CrossRef] [PubMed]
  107. Xu, J.; Xie, X.; Ye, N.; Zou, J.; Chen, H.; White, M.A.; Shi, P.Y.; Zhou, J. Design, Synthesis, and Biological Evaluation of Substituted 4,6-Dihydrospiro[[1,2,3]triazolo[4,5-b]pyridine-7,3’-indoline]-2’,5(3H)-dione Analogues as Potent NS4B Inhibitors for the Treatment of Dengue Virus Infection. J. Med. Chem. 2019, 62, 7941–7960. [Google Scholar] [CrossRef] [PubMed]
  108. Gao, Z.; Zhang, X.; Zhang, L.; Wu, S.; Ma, J.; Wang, F.; Zhou, Y.; Dai, X.; Bullitt, E.; Du, Y.; et al. A yellow fever virus NS4B inhibitor not only suppresses viral replication, but also enhances the virus activation of RIG-I-like receptor-mediated innate immune response. PLoS Pathog. 2022, 18, e1010271. [Google Scholar] [CrossRef] [PubMed]
  109. Riva, L.; Goellner, S.; Biering, S.B.; Huang, C.T.; Rubanov, A.N.; Haselmann, U.; Warnes, C.M.; De Jesus, P.D.; Martin-Sancho, L.; Terskikh, A.V.; et al. The Compound SBI-0090799 Inhibits Zika Virus Infection by Blocking De Novo Formation of the Membranous Replication Compartment. J. Virol. 2021, 95, e00996-21. [Google Scholar] [CrossRef]
  110. de Wispelaere, M.; LaCroix, A.J.; Yang, P.L. The small molecules AZD0530 and dasatinib inhibit dengue virus RNA replication via Fyn kinase. J. Virol. 2013, 87, 7367–7381. [Google Scholar] [CrossRef] [PubMed]
  111. Zmurko, J.; Neyts, J.; Dallmeier, K. Flaviviral NS4b, chameleon and jack-in-the-box roles in viral replication and pathogenesis, and a molecular target for antiviral intervention. Rev. Med. Virol. 2015, 25, 205–223. [Google Scholar] [CrossRef]
  112. Eyer, L.; Zouharova, D.; Sirmarova, J.; Fojtikova, M.; Stefanik, M.; Haviernik, J.; Nencka, R.; de Clercq, E.; Ruzek, D. Antiviral activity of the adenosine analogue BCX4430 against West Nile virus and tick-borne flaviviruses. Antivir. Res. 2017, 142, 63–67. [Google Scholar] [CrossRef]
  113. Julander, J.G.; Siddharthan, V.; Evans, J.; Taylor, R.; Tolbert, K.; Apuli, C.; Stewart, J.; Collins, P.; Gebre, M.; Neilson, S.; et al. Efficacy of the broad-spectrum antiviral compound BCX4430 against Zika virus in cell culture and in a mouse model. Antivir. Res. 2017, 137, 14–22. [Google Scholar] [CrossRef]
  114. Julander, J.G.; Demarest, J.F.; Taylor, R.; Gowen, B.B.; Walling, D.M.; Mathis, A.; Babu, Y.S. An update on the progress of galidesivir (BCX4430), a broad-spectrum antiviral. Antivir. Res. 2021, 195, 105180. [Google Scholar] [CrossRef]
  115. Escribano-Romero, E.; Jimenez de Oya, N.; Domingo, E.; Saiz, J.C. Extinction of West Nile Virus by Favipiravir through Lethal Mutagenesis. Antimicrob. Agents Chemother. 2017, 61, e01400-17. [Google Scholar] [CrossRef] [PubMed]
  116. Marlin, R.; Desjardins, D.; Contreras, V.; Lingas, G.; Solas, C.; Roques, P.; Naninck, T.; Pascal, Q.; Behillil, S.; Maisonnasse, P.; et al. Antiviral efficacy of favipiravir against Zika and SARS-CoV-2 viruses in non-human primates. Nat. Commun. 2022, 13, 5108. [Google Scholar] [CrossRef]
  117. Chen, Y.L.; Abdul Ghafar, N.; Karuna, R.; Fu, Y.; Lim, S.P.; Schul, W.; Gu, F.; Herve, M.; Yokohama, F.; Wang, G.; et al. Activation of peripheral blood mononuclear cells by dengue virus infection depotentiates balapiravir. J. Virol. 2014, 88, 1740–1747. [Google Scholar] [CrossRef] [PubMed]
  118. Deng, Y.Q.; Zhang, N.N.; Li, C.F.; Tian, M.; Hao, J.N.; Xie, X.P.; Shi, P.Y.; Qin, C.F. Adenosine Analog NITD008 Is a Potent Inhibitor of Zika Virus. Open Forum Infect. Dis. 2016, 3, ofw175. [Google Scholar] [CrossRef]
  119. Hackenbruch, C.; Maringer, Y.; Tegeler, C.M.; Walz, J.S.; Nelde, A.; Heitmann, J.S. Elevated SARS-CoV-2-Specific Antibody Levels in Patients with Post-COVID Syndrome. Viruses 2023, 15, 701. [Google Scholar] [CrossRef]
  120. Lin, K.; Good, S.S.; Julander, J.G.; Weight, A.E.; Moussa, A.; Sommadossi, J.P. AT-752, a double prodrug of a guanosine nucleotide analog, inhibits yellow fever virus in a hamster model. PLoS Negl. Trop. Dis. 2022, 16, e0009937. [Google Scholar] [CrossRef]
  121. Good, S.S.; Shannon, A.; Lin, K.; Moussa, A.; Julander, J.G.; La Colla, P.; Collu, G.; Canard, B.; Sommadossi, J.P. Evaluation of AT-752, a Double Prodrug of a Guanosine Nucleotide Analog with In Vitro and In Vivo Activity against Dengue and Other Flaviviruses. Antimicrob. Agents Chemother. 2021, 65, e00988-21. [Google Scholar] [CrossRef]
  122. Eyer, L.; Fojtikova, M.; Nencka, R.; Rudolf, I.; Hubalek, Z.; Ruzek, D. Viral RNA-Dependent RNA Polymerase Inhibitor 7-Deaza-2’-C-Methyladenosine Prevents Death in a Mouse Model of West Nile Virus Infection. Antimicrob. Agents Chemother. 2019, 63, e02093-18. [Google Scholar] [CrossRef] [PubMed]
  123. Schul, W.; Liu, W.; Xu, H.Y.; Flamand, M.; Vasudevan, S.G. A dengue fever viremia model in mice shows reduction in viral replication and suppression of the inflammatory response after treatment with antiviral drugs. J. Infect. Dis. 2007, 195, 665–674. [Google Scholar] [CrossRef] [PubMed]
  124. de Freitas, C.S.; Higa, L.M.; Sacramento, C.Q.; Ferreira, A.C.; Reis, P.A.; Delvecchio, R.; Monteiro, F.L.; Barbosa-Lima, G.; James Westgarth, H.; Vieira, Y.R.; et al. Yellow fever virus is susceptible to sofosbuvir both in vitro and in vivo. PLoS Negl. Trop. Dis. 2019, 13, e0007072. [Google Scholar] [CrossRef]
  125. Sacramento, C.Q.; de Melo, G.R.; de Freitas, C.S.; Rocha, N.; Hoelz, L.V.; Miranda, M.; Fintelman-Rodrigues, N.; Marttorelli, A.; Ferreira, A.C.; Barbosa-Lima, G.; et al. The clinically approved antiviral drug sofosbuvir inhibits Zika virus replication. Sci. Rep. 2017, 7, 40920. [Google Scholar] [CrossRef]
  126. Mesci, P.; Macia, A.; LaRock, C.N.; Tejwani, L.; Fernandes, I.R.; Suarez, N.A.; de A. Zanotto, P.M.; Beltrao-Braga, P.C.B.; Nizet, V.; Muotri, A.R. Modeling neuro-immune interactions during Zika virus infection. Hum. Mol. Genet. 2018, 27, 41–52. [Google Scholar] [CrossRef]
  127. Arora, R.; Liew, C.W.; Soh, T.S.; Otoo, D.A.; Seh, C.C.; Yue, K.; Nilar, S.; Wang, G.; Yokokawa, F.; Noble, C.G.; et al. Two RNA Tunnel Inhibitors Bind in Highly Conserved Sites in Dengue Virus NS5 Polymerase: Structural and Functional Studies. J. Virol. 2020, 94, e01130-20. [Google Scholar] [CrossRef] [PubMed]
  128. Lim, S.P.; Noble, C.G.; Seh, C.C.; Soh, T.S.; El Sahili, A.; Chan, G.K.; Lescar, J.; Arora, R.; Benson, T.; Nilar, S.; et al. Potent Allosteric Dengue Virus NS5 Polymerase Inhibitors: Mechanism of Action and Resistance Profiling. PLoS Pathog. 2016, 12, e1005737. [Google Scholar] [CrossRef] [PubMed]
  129. Stefanik, M.; Valdes, J.J.; Ezebuo, F.C.; Haviernik, J.; Uzochukwu, I.C.; Fojtikova, M.; Salat, J.; Eyer, L.; Ruzek, D. FDA-Approved Drugs Efavirenz, Tipranavir, and Dasabuvir Inhibit Replication of Multiple Flaviviruses in Vero Cells. Microorganisms 2020, 8, 599. [Google Scholar] [CrossRef] [PubMed]
  130. Kamiyama, N.; Soma, R.; Hidano, S.; Watanabe, K.; Umekita, H.; Fukuda, C.; Noguchi, K.; Gendo, Y.; Ozaki, T.; Sonoda, A.; et al. Ribavirin inhibits Zika virus (ZIKV) replication in vitro and suppresses viremia in ZIKV-infected STAT1-deficient mice. Antivir. Res. 2017, 146, 1–11. [Google Scholar] [CrossRef]
  131. Wagstaff, K.M.; Sivakumaran, H.; Heaton, S.M.; Harrich, D.; Jans, D.A. Ivermectin is a specific inhibitor of importin alpha/beta-mediated nuclear import able to inhibit replication of HIV-1 and dengue virus. Biochem. J. 2012, 443, 851–856. [Google Scholar] [CrossRef] [PubMed]
  132. Suputtamongkol, Y.; Avirutnan, P.; Mairiang, D.; Angkasekwinai, N.; Niwattayakul, K.; Yamasmith, E.; Saleh-Arong, F.A.; Songjaeng, A.; Prommool, T.; Tangthawornchaikul, N.; et al. Ivermectin Accelerates Circulating Nonstructural Protein 1 (NS1) Clearance in Adult Dengue Patients: A Combined Phase 2/3 Randomized Double-blinded Placebo Controlled Trial. Clin. Infect. Dis. 2021, 72, e586–e593. [Google Scholar] [CrossRef] [PubMed]
  133. Julander, J.G.; Furuta, Y.; Shafer, K.; Sidwell, R.W. Activity of T-1106 in a hamster model of yellow Fever virus infection. Antimicrob. Agents Chemother. 2007, 51, 1962–1966. [Google Scholar] [CrossRef] [PubMed]
  134. Khandelwal, N.; Chander, Y.; Rawat, K.D.; Riyesh, T.; Nishanth, C.; Sharma, S.; Jindal, N.; Tripathi, B.N.; Barua, S.; Kumar, N. Emetine inhibits replication of RNA and DNA viruses without generating drug-resistant virus variants. Antivir. Res. 2017, 144, 196–204. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, H.; Lao, Z.; Xu, J.; Li, Z.; Long, H.; Li, D.; Lin, L.; Liu, X.; Yu, L.; Liu, W.; et al. Antiviral activity of lycorine against Zika virus in vivo and in vitro. Virology 2020, 546, 88–97. [Google Scholar] [CrossRef] [PubMed]
  136. Yang, S.; Xu, M.; Lee, E.M.; Gorshkov, K.; Shiryaev, S.A.; He, S.; Sun, W.; Cheng, Y.S.; Hu, X.; Tharappel, A.M.; et al. Emetine inhibits Zika and Ebola virus infections through two molecular mechanisms: Inhibiting viral replication and decreasing viral entry. Cell Discov. 2018, 4, 31. [Google Scholar] [CrossRef]
  137. Pattnaik, A.; Palermo, N.; Sahoo, B.R.; Yuan, Z.; Hu, D.; Annamalai, A.S.; Vu, H.L.X.; Correas, I.; Prathipati, P.K.; Destache, C.J.; et al. Discovery of a non-nucleoside RNA polymerase inhibitor for blocking Zika virus replication through in silico screening. Antivir. Res. 2018, 151, 78–86. [Google Scholar] [CrossRef] [PubMed]
  138. Lim, S.P.; Noble, C.G.; Shi, P.Y. The dengue virus NS5 protein as a target for drug discovery. Antivir. Res. 2015, 119, 57–67. [Google Scholar] [CrossRef]
  139. Chung, K.Y.; Dong, H.; Chao, A.T.; Shi, P.Y.; Lescar, J.; Lim, S.P. Higher catalytic efficiency of N-7-methylation is responsible for processive N-7 and 2’-O methyltransferase activity in dengue virus. Virology 2010, 402, 52–60. [Google Scholar] [CrossRef] [PubMed]
  140. Noble, C.G.; Li, S.H.; Dong, H.; Chew, S.H.; Shi, P.Y. Crystal structure of dengue virus methyltransferase without S-adenosyl-L-methionine. Antivir. Res. 2014, 111, 78–81. [Google Scholar] [CrossRef]
  141. Thames, J.E.; Waters, C.D., 3rd; Valle, C.; Bassetto, M.; Aouadi, W.; Martin, B.; Selisko, B.; Falat, A.; Coutard, B.; Brancale, A.; et al. Synthesis and biological evaluation of novel flexible nucleoside analogues that inhibit flavivirus replication in vitro. Bioorg. Med. Chem. 2020, 28, 115713. [Google Scholar] [CrossRef] [PubMed]
  142. Yates, M.K.; Chatterjee, P.; Flint, M.; Arefeayne, Y.; Makuc, D.; Plavec, J.; Spiropoulou, C.F.; Seley-Radtke, K.L. Probing the Effects of Pyrimidine Functional Group Switches on Acyclic Fleximer Analogues for Antiviral Activity. Molecules 2019, 24, 3184. [Google Scholar] [CrossRef] [PubMed]
  143. Ci, Y.; Liu, Z.Y.; Zhang, N.N.; Niu, Y.; Yang, Y.; Xu, C.; Yang, W.; Qin, C.F.; Shi, L. Zika NS1-induced ER remodeling is essential for viral replication. J. Cell Biol. 2020, 219, e201903062. [Google Scholar] [CrossRef]
  144. Avirutnan, P.; Fuchs, A.; Hauhart, R.E.; Somnuke, P.; Youn, S.; Diamond, M.S.; Atkinson, J.P. Antagonism of the complement component C4 by flavivirus nonstructural protein NS1. J. Exp. Med. 2010, 207, 793–806. [Google Scholar] [CrossRef] [PubMed]
  145. Rastogi, M.; Sharma, N.; Singh, S.K. Flavivirus NS1: A multifaceted enigmatic viral protein. Virol. J. 2016, 13, 131. [Google Scholar] [CrossRef]
  146. Goldman, A.B.; Abrahams, T.G. Case report 356: Multiple findings of renal osteodystrophy reflected in the vertebral bodies and apophyseal joints of the cervical spine. Skeletal Radiol. 1986, 15, 308–312. [Google Scholar] [CrossRef] [PubMed]
  147. Rathore, A.P.; Paradkar, P.N.; Watanabe, S.; Tan, K.H.; Sung, C.; Connolly, J.E.; Low, J.; Ooi, E.E.; Vasudevan, S.G. Celgosivir treatment misfolds dengue virus NS1 protein, induces cellular pro-survival genes and protects against lethal challenge mouse model. Antivir. Res. 2011, 92, 453–460. [Google Scholar] [CrossRef] [PubMed]
  148. Barnard, T.R.; Abram, Q.H.; Lin, Q.F.; Wang, A.B.; Sagan, S.M. Molecular Determinants of Flavivirus Virion Assembly. Trends Biochem. Sci. 2021, 46, 378–390. [Google Scholar] [CrossRef]
  149. Albulescu, I.C.; Kovacikova, K.; Tas, A.; Snijder, E.J.; van Hemert, M.J. Suramin inhibits Zika virus replication by interfering with virus attachment and release of infectious particles. Antivir. Res. 2017, 143, 230–236. [Google Scholar] [CrossRef]
  150. Klaitong, P.; Smith, D.R. Roles of Non-Structural Protein 4A in Flavivirus Infection. Viruses 2021, 13, 2077. [Google Scholar] [CrossRef]
  151. Miller, S.; Kastner, S.; Krijnse-Locker, J.; Buhler, S.; Bartenschlager, R. The non-structural protein 4A of dengue virus is an integral membrane protein inducing membrane alterations in a 2K-regulated manner. J. Biol. Chem. 2007, 282, 8873–8882. [Google Scholar] [CrossRef] [PubMed]
  152. Wang, Y.; Xie, X.; Shi, P.Y. Flavivirus NS4B protein: Structure, function, and antiviral discovery. Antiviral Res. 2022, 207, 105423. [Google Scholar] [CrossRef]
  153. Chatel-Chaix, L.; Fischl, W.; Scaturro, P.; Cortese, M.; Kallis, S.; Bartenschlager, M.; Fischer, B.; Bartenschlager, R. A Combined Genetic-Proteomic Approach Identifies Residues within Dengue Virus NS4B Critical for Interaction with NS3 and Viral Replication. J. Virol. 2015, 89, 7170–7186. [Google Scholar] [CrossRef] [PubMed]
  154. Goethals, O.; Kaptein, S.J.F.; Kesteleyn, B.; Bonfanti, J.F.; Van Wesenbeeck, L.; Bardiot, D.; Verschoor, E.J.; Verstrepen, B.E.; Fagrouch, Z.; Putnak, J.R.; et al. Blocking NS3-NS4B interaction inhibits dengue virus in non-human primates. Nature 2023, 615, 678–686. [Google Scholar] [CrossRef]
  155. Ackaert, O.; Vanhoutte, F.; Verpoorten, N.; Buelens, A.; Lachau-Durand, S.; Lammens, L.; Hoetelmans, R.; Van Loock, M.; Herrera-Taracena, G. Safety, Tolerability, and Pharmacokinetics of JNJ-1802, a Pan-serotype Dengue Direct Antiviral Small Molecule, in a Phase 1, Double-Blind, Randomized, Dose-Escalation Study in Healthy Volunteers. Clin. Infect. Dis. 2023, 77, 857–865. [Google Scholar] [CrossRef] [PubMed]
  156. Zhou, Y.; Ray, D.; Zhao, Y.; Dong, H.; Ren, S.; Li, Z.; Guo, Y.; Bernard, K.A.; Shi, P.Y.; Li, H. Structure and function of flavivirus NS5 methyltransferase. J. Virol. 2007, 81, 3891–3903. [Google Scholar] [CrossRef]
  157. Geraghty, R.J.; Aliota, M.T.; Bonnac, L.F. Broad-Spectrum Antiviral Strategies and Nucleoside Analogues. Viruses 2021, 13, 667. [Google Scholar] [CrossRef] [PubMed]
  158. Abdullah Al Awadh, A. Nucleotide and nucleoside-based drugs: Past, present, and future. Saudi J. Biol. Sci. 2022, 29, 103481. [Google Scholar] [CrossRef] [PubMed]
  159. Eyer, L.; Nencka, R.; de Clercq, E.; Seley-Radtke, K.; Ruzek, D. Nucleoside analogs as a rich source of antiviral agents active against arthropod-borne flaviviruses. Antivir. Chem. Chemother. 2018, 26, 2040206618761299. [Google Scholar] [CrossRef] [PubMed]
  160. Kataev, V.E.; Garifullin, B.F. Antiviral nucleoside analogs. Chem. Heterocycl. Compd. 2021, 57, 326–341. [Google Scholar] [CrossRef] [PubMed]
  161. Stedman, C. Sofosbuvir, a NS5B polymerase inhibitor in the treatment of hepatitis C: A review of its clinical potential. Therap. Adv. Gastroenterol. 2014, 7, 131–140. [Google Scholar] [CrossRef] [PubMed]
  162. Alvarez, D.M.; Castillo, E.; Duarte, L.F.; Arriagada, J.; Corrales, N.; Farias, M.A.; Henriquez, A.; Agurto-Munoz, C.; Gonzalez, P.A. Current Antivirals and Novel Botanical Molecules Interfering With Herpes Simplex Virus Infection. Front. Microbiol. 2020, 11, 139. [Google Scholar] [CrossRef] [PubMed]
  163. Yin, Z.; Chen, Y.L.; Schul, W.; Wang, Q.Y.; Gu, F.; Duraiswamy, J.; Kondreddi, R.R.; Niyomrattanakit, P.; Lakshminarayana, S.B.; Goh, A.; et al. An adenosine nucleoside inhibitor of dengue virus. Proc. Natl. Acad. Sci. USA 2009, 106, 20435–20439. [Google Scholar] [CrossRef] [PubMed]
  164. McMahon, J.H.; Lau, J.S.Y.; Coldham, A.; Roney, J.; Hagenauer, M.; Price, S.; Bryant, M.; Garlick, J.; Paterson, A.; Lee, S.J.; et al. Favipiravir in early symptomatic COVID-19, a randomised placebo-controlled trial. eClinicalMedicine 2022, 54, 101703. [Google Scholar] [CrossRef] [PubMed]
  165. Guedj, J.; Piorkowski, G.; Jacquot, F.; Madelain, V.; Nguyen, T.H.T.; Rodallec, A.; Gunther, S.; Carbonnelle, C.; Mentre, F.; Raoul, H.; et al. Antiviral efficacy of favipiravir against Ebola virus: A translational study in cynomolgus macaques. PLoS Med. 2018, 15, e1002535. [Google Scholar] [CrossRef] [PubMed]
  166. Franco, E.J.; Cella, E.; Tao, X.; Hanrahan, K.C.; Azarian, T.; Brown, A.N. Favipiravir Suppresses Zika Virus (ZIKV) through Activity as a Mutagen. Microorganisms 2023, 11, 1342. [Google Scholar] [CrossRef] [PubMed]
  167. Karuna, R.; Yokokawa, F.; Wang, K.; Zhang, J.; Xu, H.; Wang, G.; Ding, M.; Chan, W.L.; Abdul Ghafar, N.; Leonardi, A.; et al. A Cyclic Phosphoramidate Prodrug of 2’-Deoxy-2’-Fluoro-2’-C-Methylguanosine for the Treatment of Dengue Virus Infection. Antimicrob. Agents Chemother. 2020, 64, e00654-20. [Google Scholar] [CrossRef]
  168. Toniutto, P.; Fabris, C.; Bitetto, D.; Fumolo, E.; Fornasiere, E.; Pirisi, M. R-1626, a specific oral NS5B polymerase inhibitor of hepatitis C virus. IDrugs 2008, 11, 738–749. [Google Scholar] [PubMed]
  169. Nguyen, N.M.; Tran, C.N.; Phung, L.K.; Duong, K.T.; Huynh Hle, A.; Farrar, J.; Nguyen, Q.T.; Tran, H.T.; Nguyen, C.V.; Merson, L.; et al. A randomized, double-blind placebo controlled trial of balapiravir, a polymerase inhibitor, in adult dengue patients. J. Infect. Dis. 2013, 207, 1442–1450. [Google Scholar] [CrossRef]
  170. Lee, M.F.; Wu, Y.S.; Poh, C.L. Molecular Mechanisms of Antiviral Agents against Dengue Virus. Viruses 2023, 15, 705. [Google Scholar] [CrossRef] [PubMed]
  171. Arnold, J.J.; Sharma, S.D.; Feng, J.Y.; Ray, A.S.; Smidansky, E.D.; Kireeva, M.L.; Cho, A.; Perry, J.; Vela, J.E.; Park, Y.; et al. Sensitivity of mitochondrial transcription and resistance of RNA polymerase II dependent nuclear transcription to antiviral ribonucleosides. PLoS Pathog. 2012, 8, e1003030. [Google Scholar] [CrossRef]
  172. Lim, S.P.; Wang, Q.Y.; Noble, C.G.; Chen, Y.L.; Dong, H.; Zou, B.; Yokokawa, F.; Nilar, S.; Smith, P.; Beer, D.; et al. Ten years of dengue drug discovery: Progress and prospects. Antivir. Res. 2013, 100, 500–519. [Google Scholar] [CrossRef]
  173. Dragoni, F.; Boccuto, A.; Picarazzi, F.; Giannini, A.; Giammarino, F.; Saladini, F.; Mori, M.; Mastrangelo, E.; Zazzi, M.; Vicenti, I. Evaluation of sofosbuvir activity and resistance profile against West Nile virus in vitro. Antivir. Res. 2020, 175, 104708. [Google Scholar] [CrossRef] [PubMed]
  174. Xu, H.T.; Colby-Germinario, S.P.; Hassounah, S.A.; Fogarty, C.; Osman, N.; Palanisamy, N.; Han, Y.; Oliveira, M.; Quan, Y.; Wainberg, M.A. Evaluation of Sofosbuvir (beta-D-2’-deoxy-2’-alpha-fluoro-2’-beta-C-methyluridine) as an inhibitor of Dengue virus replication. Sci. Rep. 2017, 7, 6345. [Google Scholar] [CrossRef]
  175. Appleby, T.C.; Perry, J.K.; Murakami, E.; Barauskas, O.; Feng, J.; Cho, A.; Fox, D., 3rd; Wetmore, D.R.; McGrath, M.E.; Ray, A.S.; et al. Viral replication. Structural basis for RNA replication by the hepatitis C virus polymerase. Science 2015, 347, 771–775. [Google Scholar] [CrossRef] [PubMed]
  176. Rattanaburee, T.; Junking, M.; Panya, A.; Sawasdee, N.; Songprakhon, P.; Suttitheptumrong, A.; Limjindaporn, T.; Haegeman, G.; Yenchitsomanus, P.T. Inhibition of dengue virus production and cytokine/chemokine expression by ribavirin and compound A. Antivir. Res. 2015, 124, 83–92. [Google Scholar] [CrossRef]
  177. Noble, C.G.; Lim, S.P.; Chen, Y.L.; Liew, C.W.; Yap, L.; Lescar, J.; Shi, P.Y. Conformational flexibility of the Dengue virus RNA-dependent RNA polymerase revealed by a complex with an inhibitor. J. Virol. 2013, 87, 5291–5295. [Google Scholar] [CrossRef] [PubMed]
  178. Niyomrattanakit, P.; Chen, Y.L.; Dong, H.; Yin, Z.; Qing, M.; Glickman, J.F.; Lin, K.; Mueller, D.; Voshol, H.; Lim, J.Y.; et al. Inhibition of dengue virus polymerase by blocking of the RNA tunnel. J. Virol. 2010, 84, 5678–5686. [Google Scholar] [CrossRef] [PubMed]
  179. Pryor, M.J.; Rawlinson, S.M.; Butcher, R.E.; Barton, C.L.; Waterhouse, T.A.; Vasudevan, S.G.; Bardin, P.G.; Wright, P.J.; Jans, D.A.; Davidson, A.D. Nuclear localization of dengue virus nonstructural protein 5 through its importin alpha/beta-recognized nuclear localization sequences is integral to viral infection. Traffic 2007, 8, 795–807. [Google Scholar] [CrossRef] [PubMed]
  180. Perera, R.; Riley, C.; Isaac, G.; Hopf-Jannasch, A.S.; Moore, R.J.; Weitz, K.W.; Pasa-Tolic, L.; Metz, T.O.; Adamec, J.; Kuhn, R.J. Dengue virus infection perturbs lipid homeostasis in infected mosquito cells. PLoS Pathog. 2012, 8, e1002584. [Google Scholar] [CrossRef]
  181. Heaton, N.S.; Randall, G. Dengue virus-induced autophagy regulates lipid metabolism. Cell Host Microbe 2010, 8, 422–432. [Google Scholar] [CrossRef] [PubMed]
  182. Samsa, M.M.; Mondotte, J.A.; Iglesias, N.G.; Assuncao-Miranda, I.; Barbosa-Lima, G.; Da Poian, A.T.; Bozza, P.T.; Gamarnik, A.V. Dengue virus capsid protein usurps lipid droplets for viral particle formation. PLoS Pathog. 2009, 5, e1000632. [Google Scholar] [CrossRef] [PubMed]
  183. Nicholls, C.M.R.; Sevvana, M.; Kuhn, R.J. Structure-guided paradigm shifts in flavivirus assembly and maturation mechanisms. Adv. Virus Res. 2020, 108, 33–83. [Google Scholar] [CrossRef]
  184. Chambers, T.J.; Hahn, C.S.; Galler, R.; Rice, C.M. Flavivirus genome organization, expression, and replication. Annu. Rev. Microbiol. 1990, 44, 649–688. [Google Scholar] [CrossRef] [PubMed]
  185. Crabtree, M.B.; Kinney, R.M.; Miller, B.R. Deglycosylation of the NS1 protein of dengue 2 virus, strain 16681: Construction and characterization of mutant viruses. Arch. Virol. 2005, 150, 771–786. [Google Scholar] [CrossRef] [PubMed]
  186. Helenius, A.; Aebi, M. Intracellular functions of N-linked glycans. Science 2001, 291, 2364–2369. [Google Scholar] [CrossRef] [PubMed]
  187. Whitby, K.; Pierson, T.C.; Geiss, B.; Lane, K.; Engle, M.; Zhou, Y.; Doms, R.W.; Diamond, M.S. Castanospermine, a potent inhibitor of dengue virus infection in vitro and in vivo. J. Virol. 2005, 79, 8698–8706. [Google Scholar] [CrossRef]
  188. Watanabe, S.; Chan, K.W.; Dow, G.; Ooi, E.E.; Low, J.G.; Vasudevan, S.G. Optimizing celgosivir therapy in mouse models of dengue virus infection of serotypes 1 and 2: The search for a window for potential therapeutic efficacy. Antivir. Res. 2016, 127, 10–19. [Google Scholar] [CrossRef] [PubMed]
  189. Low, J.G.; Sung, C.; Wijaya, L.; Wei, Y.; Rathore, A.P.S.; Watanabe, S.; Tan, B.H.; Toh, L.; Chua, L.T.; Hou, Y.; et al. Efficacy and safety of celgosivir in patients with dengue fever (CELADEN): A phase 1b, randomised, double-blind, placebo-controlled, proof-of-concept trial. Lancet Infect. Dis. 2014, 14, 706–715. [Google Scholar] [CrossRef]
  190. Callahan, M.; Treston, A.M.; Lin, G.; Smith, M.; Kaufman, B.; Khaliq, M.; Evans DeWald, L.; Spurgers, K.; Warfield, K.L.; Lowe, P.; et al. Randomized single oral dose phase 1 study of safety, tolerability, and pharmacokinetics of Iminosugar UV-4 Hydrochloride (UV-4B) in healthy subjects. PLoS Negl. Trop. Dis. 2022, 16, e0010636. [Google Scholar] [CrossRef]
  191. Chang, J.; Schul, W.; Butters, T.D.; Yip, A.; Liu, B.; Goh, A.; Lakshminarayana, S.B.; Alonzi, D.; Reinkensmeier, G.; Pan, X.; et al. Combination of alpha-glucosidase inhibitor and ribavirin for the treatment of dengue virus infection in vitro and in vivo. Antivir. Res. 2011, 89, 26–34. [Google Scholar] [CrossRef]
  192. Evans DeWald, L.; Starr, C.; Butters, T.; Treston, A.; Warfield, K.L. Iminosugars: A host-targeted approach to combat Flaviviridae infections. Antivir. Res. 2020, 184, 104881. [Google Scholar] [CrossRef] [PubMed]
  193. Warfield, K.L.; Plummer, E.; Alonzi, D.S.; Wolfe, G.W.; Sampath, A.; Nguyen, T.; Butters, T.D.; Enterlein, S.G.; Stavale, E.J.; Shresta, S.; et al. A Novel Iminosugar UV-12 with Activity against the Diverse Viruses Influenza and Dengue (Novel Iminosugar Antiviral for Influenza and Dengue). Viruses 2015, 7, 2404–2427. [Google Scholar] [CrossRef]
  194. Merino-Ramos, T.; Vazquez-Calvo, A.; Casas, J.; Sobrino, F.; Saiz, J.C.; Martin-Acebes, M.A. Modification of the Host Cell Lipid Metabolism Induced by Hypolipidemic Drugs Targeting the Acetyl Coenzyme A Carboxylase Impairs West Nile Virus Replication. Antimicrob. Agents Chemother. 2016, 60, 307–315. [Google Scholar] [CrossRef]
  195. Anwar, A.; Hosoya, T.; Leong, K.M.; Onogi, H.; Okuno, Y.; Hiramatsu, T.; Koyama, H.; Suzuki, M.; Hagiwara, M.; Garcia-Blanco, M.A. The kinase inhibitor SFV785 dislocates dengue virus envelope protein from the replication complex and blocks virus assembly. PLoS ONE 2011, 6, e23246. [Google Scholar] [CrossRef] [PubMed]
  196. Blazquez, A.B.; Saiz, J.C. Potential for Protein Kinase Pharmacological Regulation in Flaviviridae Infections. Int. J. Mol. Sci. 2020, 21, 9524. [Google Scholar] [CrossRef]
  197. Martinez-Gutierrez, M.; Correa-Londono, L.A.; Castellanos, J.E.; Gallego-Gomez, J.C.; Osorio, J.E. Lovastatin delays infection and increases survival rates in AG129 mice infected with dengue virus serotype 2. PLoS ONE 2014, 9, e87412. [Google Scholar] [CrossRef] [PubMed]
  198. Whitehorn, J.; Nguyen, C.V.V.; Khanh, L.P.; Kien, D.T.H.; Quyen, N.T.H.; Tran, N.T.T.; Hang, N.T.; Truong, N.T.; Hue Tai, L.T.; Cam Huong, N.T.; et al. Lovastatin for the Treatment of Adult Patients With Dengue: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Infect. Dis. 2016, 62, 468–476. [Google Scholar] [CrossRef]
  199. Sung, C.; Wei, Y.; Watanabe, S.; Lee, H.S.; Khoo, Y.M.; Fan, L.; Rathore, A.P.; Chan, K.W.; Choy, M.M.; Kamaraj, U.S.; et al. Extended Evaluation of Virological, Immunological and Pharmacokinetic Endpoints of CELADEN: A Randomized, Placebo-Controlled Trial of Celgosivir in Dengue Fever Patients. PLoS Negl. Trop. Dis. 2016, 10, e0004851. [Google Scholar] [CrossRef] [PubMed]
  200. Rothwell, C.; Lebreton, A.; Young Ng, C.; Lim, J.Y.; Liu, W.; Vasudevan, S.; Labow, M.; Gu, F.; Gaither, L.A. Cholesterol biosynthesis modulation regulates dengue viral replication. Virology 2009, 389, 8–19. [Google Scholar] [CrossRef] [PubMed]
  201. Osuna-Ramos, J.F.; Farfan-Morales, C.N.; Cordero-Rivera, C.D.; De Jesus-Gonzalez, L.A.; Reyes-Ruiz, J.M.; Hurtado-Monzon, A.M.; Palacios-Rapalo, S.N.; Jimenez-Camacho, R.; Meraz-Rios, M.A.; Del Angel, R.M. Cholesterol-Lowering Drugs as Potential Antivirals: A Repurposing Approach against Flavivirus Infections. Viruses 2023, 15, 1465. [Google Scholar] [CrossRef] [PubMed]
  202. Smith, J.L.; Sheridan, K.; Parkins, C.J.; Frueh, L.; Jemison, A.L.; Strode, K.; Dow, G.; Nilsen, A.; Hirsch, A.J. Characterization and structure-activity relationship analysis of a class of antiviral compounds that directly bind dengue virus capsid protein and are incorporated into virions. Antivir. Res. 2018, 155, 12–19. [Google Scholar] [CrossRef] [PubMed]
  203. Xia, H.; Xie, X.; Zou, J.; Noble, C.G.; Russell, W.K.; Holthauzen, L.M.F.; Choi, K.H.; White, M.A.; Shi, P.Y. A cocrystal structure of dengue capsid protein in complex of inhibitor. Proc. Natl. Acad. Sci. USA 2020, 117, 17992–18001. [Google Scholar] [CrossRef] [PubMed]
  204. Byrd, C.M.; Dai, D.; Grosenbach, D.W.; Berhanu, A.; Jones, K.F.; Cardwell, K.B.; Schneider, C.; Wineinger, K.A.; Page, J.M.; Harver, C.; et al. A novel inhibitor of dengue virus replication that targets the capsid protein. Antimicrob. Agents Chemother. 2013, 57, 15–25. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, S.H.; Syu, W.J.; Hu, S.T. Identification of the homotypic interaction domain of the core protein of dengue virus type 2. J. Gen. Virol. 2004, 85, 2307–2314. [Google Scholar] [CrossRef]
  206. Byk, L.A.; Gamarnik, A.V. Properties and Functions of the Dengue Virus Capsid Protein. Annu. Rev. Virol. 2016, 3, 263–281. [Google Scholar] [CrossRef]
  207. Sotcheff, S.; Routh, A. Understanding Flavivirus Capsid Protein Functions: The Tip of the Iceberg. Pathogens 2020, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  208. Boonyasuppayakorn, S.; Reichert, E.D.; Manzano, M.; Nagarajan, K.; Padmanabhan, R. Amodiaquine, an antimalarial drug, inhibits dengue virus type 2 replication and infectivity. Antivir. Res. 2014, 106, 125–134. [Google Scholar] [CrossRef] [PubMed]
  209. Cheung, Y.Y.; Chen, K.C.; Chen, H.; Seng, E.K.; Chu, J.J. Antiviral activity of lanatoside C against dengue virus infection. Antivir. Res. 2014, 111, 93–99. [Google Scholar] [CrossRef]
  210. Kato, F.; Ishida, Y.; Oishi, S.; Fujii, N.; Watanabe, S.; Vasudevan, S.G.; Tajima, S.; Takasaki, T.; Suzuki, Y.; Ichiyama, K.; et al. Novel antiviral activity of bromocriptine against dengue virus replication. Antivir. Res. 2016, 131, 141–147. [Google Scholar] [CrossRef]
  211. Zakaria, M.K.; Carletti, T.; Marcello, A. Cellular Targets for the Treatment of Flavivirus Infections. Front. Cell. Infect. Microbiol. 2018, 8, 398. [Google Scholar] [CrossRef]
  212. Scroggs, S.L.P.; Andrade, C.C.; Chinnasamy, R.; Azar, S.R.; Schirtzinger, E.E.; Garcia, E.I.; Arterburn, J.B.; Hanley, K.A.; Rossi, S.L. Old Drugs with New Tricks: Efficacy of Fluoroquinolones to Suppress Replication of Flaviviruses. Viruses 2020, 12, 1022. [Google Scholar] [CrossRef] [PubMed]
  213. Haviernik, J.; Stefanik, M.; Fojtikova, M.; Kali, S.; Tordo, N.; Rudolf, I.; Hubalek, Z.; Eyer, L.; Ruzek, D. Arbidol (Umifenovir): A Broad-Spectrum Antiviral Drug That Inhibits Medically Important Arthropod-Borne Flaviviruses. Viruses 2018, 10, 184. [Google Scholar] [CrossRef]
  214. Puschnik, A.S.; Marceau, C.D.; Ooi, Y.S.; Majzoub, K.; Rinis, N.; Contessa, J.N.; Carette, J.E. A Small-Molecule Oligosaccharyltransferase Inhibitor with Pan-flaviviral Activity. Cell Rep. 2017, 21, 3032–3039. [Google Scholar] [CrossRef] [PubMed]
  215. Othman, M.A.; Abou-Donia, M.B. Pharmacokinetic profile of (+/−)-gossypol in male Sprague-Dawley rats following single intravenous and oral and subchronic oral administration. Proc. Soc. Exp. Biol. Med. 1988, 188, 17–22. [Google Scholar] [CrossRef] [PubMed]
  216. Eyre, N.S.; Kirby, E.N.; Anfiteatro, D.R.; Bracho, G.; Russo, A.G.; White, P.A.; Aloia, A.L.; Beard, M.R. Identification of Estrogen Receptor Modulators as Inhibitors of Flavivirus Infection. Antimicrob. Agents Chemother. 2020, 64, e00289. [Google Scholar] [CrossRef]
  217. Quintana, V.M.; Selisko, B.; Brunetti, J.E.; Eydoux, C.; Guillemot, J.C.; Canard, B.; Damonte, E.B.; Julander, J.G.; Castilla, V. Antiviral activity of the natural alkaloid anisomycin against dengue and Zika viruses. Antivir. Res 2020, 176, 104749. [Google Scholar] [CrossRef] [PubMed]
  218. Carocci, M.; Hinshaw, S.M.; Rodgers, M.A.; Villareal, V.A.; Burri, D.J.; Pilankatta, R.; Maharaj, N.P.; Gack, M.U.; Stavale, E.J.; Warfield, K.L.; et al. The bioactive lipid 4-hydroxyphenyl retinamide inhibits flavivirus replication. Antimicrob. Agents Chemother. 2015, 59, 85–95. [Google Scholar] [CrossRef]
  219. Zhou, T.; Tan, L.; Cederquist, G.Y.; Fan, Y.; Hartley, B.J.; Mukherjee, S.; Tomishima, M.; Brennand, K.J.; Zhang, Q.; Schwartz, R.E.; et al. High-Content Screening in hPSC-Neural Progenitors Identifies Drug Candidates that Inhibit Zika Virus Infection in Fetal-like Organoids and Adult Brain. Cell Stem Cell 2017, 21, 274–283.e5. [Google Scholar] [CrossRef] [PubMed]
  220. Heiss, B.L.; Maximova, O.A.; Pletnev, A.G. Insertion of microRNA targets into the flavivirus genome alters its highly neurovirulent phenotype. J. Virol. 2011, 85, 1464–1472. [Google Scholar] [CrossRef]
  221. Heiss, B.L.; Maximova, O.A.; Thach, D.C.; Speicher, J.M.; Pletnev, A.G. MicroRNA targeting of neurotropic flavivirus: Effective control of virus escape and reversion to neurovirulent phenotype. J. Virol. 2012, 86, 5647–5659. [Google Scholar] [CrossRef]
  222. Yen, L.C.; Lin, Y.L.; Sung, H.H.; Liao, J.T.; Tsao, C.H.; Su, C.M.; Lin, C.K.; Liao, C.L. Neurovirulent flavivirus can be attenuated in mice by incorporation of neuron-specific microRNA recognition elements into viral genome. Vaccine 2013, 31, 5915–5922. [Google Scholar] [CrossRef]
  223. Sharma, H.; Tripathi, A.; Kumari, B.; Vrati, S.; Banerjee, A. Artificial MicroRNA-Mediated Inhibition of Japanese Encephalitis Virus Replication in Neuronal Cells. Nucleic Acid Ther. 2018, 28, 357–365. [Google Scholar] [CrossRef]
  224. Karothia, D.; Kumar Dash, P.; Parida, M.; Bhagyawant, S.S.; Kumar, J.S. Vector derived artificial miRNA mediated inhibition of West Nile virus replication and protein expression. Gene 2020, 729, 144300. [Google Scholar] [CrossRef] [PubMed]
  225. Xie, P.W.; Xie, Y.; Zhang, X.J.; Huang, H.; He, L.N.; Wang, X.J.; Wang, S.Q. Inhibition of Dengue virus 2 replication by artificial micrornas targeting the conserved regions. Nucleic Acid Ther. 2013, 23, 244–252. [Google Scholar] [CrossRef] [PubMed]
  226. Shen, T.; Liu, K.; Miao, D.; Cao, R.; Chen, P. Effective inhibition of Japanese encephalitis virus replication by shRNAs targeting various viral genes in vitro and in vivo. Virology 2014, 454–455, 48–59. [Google Scholar] [CrossRef] [PubMed]
  227. Wu, Z.; Xue, Y.; Wang, B.; Du, J.; Jin, Q. Broad-spectrum antiviral activity of RNA interference against four genotypes of Japanese encephalitis virus based on single microRNA polycistrons. PLoS ONE 2011, 6, e26304. [Google Scholar] [CrossRef]
  228. Brostoff, T.; Pesavento, P.A.; Barker, C.M.; Kenney, J.L.; Dietrich, E.A.; Duggal, N.K.; Bosco-Lauth, A.M.; Brault, A.C. MicroRNA reduction of neuronal West Nile virus replication attenuates and affords a protective immune response in mice. Vaccine 2016, 34, 5366–5375. [Google Scholar] [CrossRef]
  229. Natekar, J.P.; Rothan, H.A.; Arora, K.; Strate, P.G.; Kumar, M. Cellular microRNA-155 Regulates Virus-Induced Inflammatory Response and Protects against Lethal West Nile Virus Infection. Viruses 2019, 12, 9. [Google Scholar] [CrossRef] [PubMed]
  230. Pacca, C.C.; Severino, A.A.; Mondini, A.; Rahal, P.; D’Avila, S.G.; Cordeiro, J.A.; Nogueira, M.C.; Bronzoni, R.V.; Nogueira, M.L. RNA interference inhibits yellow fever virus replication in vitro and in vivo. Virus Genes 2009, 38, 224–231. [Google Scholar] [CrossRef] [PubMed]
  231. Zhang, R.; Fu, Y.; Cheng, M.; Ma, W.; Zheng, N.; Wang, Y.; Wu, Z. sEVs(RVG) selectively delivers antiviral siRNA to fetus brain, inhibits ZIKV infection and mitigates ZIKV-induced microcephaly in mouse model. Mol. Ther. 2022, 30, 2078–2091. [Google Scholar] [CrossRef] [PubMed]
  232. Chatterjee, S.; Kordbacheh, R.; Sin, J. Extracellular Vesicles: A Novel Mode of Viral Propagation Exploited by Enveloped and Non-Enveloped Viruses. Microorganisms 2024, 12, 274. [Google Scholar] [CrossRef] [PubMed]
  233. El Safadi, D.; Mokhtari, A.; Krejbich, M.; Lagrave, A.; Hirigoyen, U.; Lebeau, G.; Viranaicken, W.; Krejbich-Trotot, P. Exosome-Mediated Antigen Delivery: Unveiling Novel Strategies in Viral Infection Control and Vaccine Design. Vaccines 2024, 12, 280. [Google Scholar] [CrossRef]
  234. Dai, Z.; Cai, R.; Zeng, H.; Zhu, H.; Dou, Y.; Sun, S. Exosome may be the next generation of promising cell-free vaccines. Hum. Vaccin. Immunother. 2024, 20, 2345940. [Google Scholar] [CrossRef] [PubMed]
  235. Latanova, A.; Karpov, V.; Starodubova, E. Extracellular Vesicles in Flaviviridae Pathogenesis: Their Roles in Viral Transmission, Immune Evasion, and Inflammation. Int. J. Mol. Sci. 2024, 25, 2144. [Google Scholar] [CrossRef]
  236. Ishikawa, T.; Narita, K.; Matsuyama, K.; Masuda, M. Dissemination of the Flavivirus Subgenomic Replicon Genome and Viral Proteins by Extracellular Vesicles. Viruses 2024, 16, 524. [Google Scholar] [CrossRef]
  237. Martins, S.T.; Kuczera, D.; Lotvall, J.; Bordignon, J.; Alves, L.R. Characterization of Dendritic Cell-Derived Extracellular Vesicles During Dengue Virus Infection. Front. Microbiol. 2018, 9, 1792. [Google Scholar] [CrossRef] [PubMed]
  238. Slonchak, A.; Clarke, B.; Mackenzie, J.; Amarilla, A.A.; Setoh, Y.X.; Khromykh, A.A. West Nile virus infection and interferon alpha treatment alter the spectrum and the levels of coding and noncoding host RNAs secreted in extracellular vesicles. BMC Genom. 2019, 20, 474. [Google Scholar] [CrossRef] [PubMed]
  239. Block, L.N.; Schmidt, J.K.; Keuler, N.S.; McKeon, M.C.; Bowman, B.D.; Wiepz, G.J.; Golos, T.G. Zika virus impacts extracellular vesicle composition and cellular gene expression in macaque early gestation trophoblasts. Sci. Rep. 2022, 12, 7348. [Google Scholar] [CrossRef] [PubMed]
  240. Huang, L.; Li, H.; Ye, Z.; Xu, Q.; Fu, Q.; Sun, W.; Qi, W.; Yue, J. Berbamine inhibits Japanese encephalitis virus (JEV) infection by compromising TPRMLs-mediated endolysosomal trafficking of low-density lipoprotein receptor (LDLR). Emerg. Microbes Infect. 2021, 10, 1257–1271. [Google Scholar] [CrossRef]
  241. Conzelmann, C.; Gross, R.; Zou, M.; Kruger, F.; Gorgens, A.; Gustafsson, M.O.; El Andaloussi, S.; Munch, J.; Muller, J.A. Salivary extracellular vesicles inhibit Zika virus but not SARS-CoV-2 infection. J. Extracell. Vesicles 2020, 9, 1808281. [Google Scholar] [CrossRef]
  242. Li, S.; Zhu, A.; Ren, K.; Li, S.; Chen, L. DEFA1B inhibits ZIKV replication and retards cell cycle progression through interaction with ORC1. Life Sci. 2020, 263, 118564. [Google Scholar] [CrossRef] [PubMed]
  243. Wang, C.; Li, M.; Xia, X.; Fu, Y.; Wang, Y.; Xu, W.; Wei, H.; Wei, L. Construction of exosome-loaded LL-37 and its protection against zika virus infection. Antivir. Res. 2024, 225, 105855. [Google Scholar] [CrossRef] [PubMed]
  244. Zou, X.; Yuan, M.; Zhang, T.; Zheng, N.; Wu, Z. EVs Containing Host Restriction Factor IFITM3 Inhibited ZIKV Infection of Fetuses in Pregnant Mice through Trans-placenta Delivery. Mol. Ther. 2021, 29, 176–190. [Google Scholar] [CrossRef] [PubMed]
  245. Yenuganti, V.R.; Afroz, S.; Khan, R.A.; Bharadwaj, C.; Nabariya, D.K.; Nayak, N.; Subbiah, M.; Chintala, K.; Banerjee, S.; Reddanna, P.; et al. Milk exosomes elicit a potent anti-viral activity against dengue virus. J. Nanobiotechnology 2022, 20, 317. [Google Scholar] [CrossRef]
  246. Zhang, Y.; Dai, Y.; Wang, J.; Xu, Y.; Li, Z.; Lu, J.; Xu, Y.; Zhong, J.; Ding, S.W.; Li, Y. Mouse circulating extracellular vesicles contain virus-derived siRNAs active in antiviral immunity. EMBO J. 2022, 41, e109902. [Google Scholar] [CrossRef]
  247. Ahmed, W.; Neelakanta, G.; Sultana, H. Tetraspanins as Potential Therapeutic Candidates for Targeting Flaviviruses. Front. Immunol. 2021, 12, 630571. [Google Scholar] [CrossRef]
  248. Santos, P.; Almeida, F. Exosome-Based Vaccines: History, Current State, and Clinical Trials. Front. Immunol. 2021, 12, 711565. [Google Scholar] [CrossRef] [PubMed]
  249. Aliota, M.T.; Caine, E.A.; Walker, E.C.; Larkin, K.E.; Camacho, E.; Osorio, J.E. Characterization of Lethal Zika Virus Infection in AG129 Mice. PLoS Negl. Trop. Dis. 2016, 10, e0004682. [Google Scholar] [CrossRef] [PubMed]
  250. Lazear, H.M.; Govero, J.; Smith, A.M.; Platt, D.J.; Fernandez, E.; Miner, J.J.; Diamond, M.S. A Mouse Model of Zika Virus Pathogenesis. Cell Host Microbe 2016, 19, 720–730. [Google Scholar] [CrossRef] [PubMed]
  251. Kayesh, M.E.H.; Tsukiyama-Kohara, K. Mammalian animal models for dengue virus infection: A recent overview. Arch. Virol. 2022, 167, 31–44. [Google Scholar] [CrossRef]
  252. Alves Dos Santos, E.; Fink, K. Animal Models for Dengue and Zika Vaccine Development. Adv. Exp. Med. Biol. 2018, 1062, 215–239. [Google Scholar] [CrossRef] [PubMed]
  253. Tripathi, A.; Banerjee, A.; Vrati, S. Development and characterization of an animal model of Japanese encephalitis virus infection in adolescent C57BL/6 mouse. Dis. Model Mech. 2021, 14, dmm.049176. [Google Scholar] [CrossRef]
  254. Calvert, A.E.; Dixon, K.L.; Delorey, M.J.; Blair, C.D.; Roehrig, J.T. Development of a small animal peripheral challenge model of Japanese encephalitis virus using interferon deficient AG129 mice and the SA14-14-2 vaccine virus strain. Vaccine 2014, 32, 258–264. [Google Scholar] [CrossRef] [PubMed]
  255. Zhu, B.; Ye, J.; Nie, Y.; Ashraf, U.; Zohaib, A.; Duan, X.; Fu, Z.F.; Song, Y.; Chen, H.; Cao, S. MicroRNA-15b Modulates Japanese Encephalitis Virus-Mediated Inflammation via Targeting RNF125. J. Immunol. 2015, 195, 2251–2262. [Google Scholar] [CrossRef] [PubMed]
  256. Meier, K.C.; Gardner, C.L.; Khoretonenko, M.V.; Klimstra, W.B.; Ryman, K.D. A mouse model for studying viscerotropic disease caused by yellow fever virus infection. PLoS Pathog. 2009, 5, e1000614. [Google Scholar] [CrossRef]
  257. Monath, T.P. Treatment of yellow fever. Antivir. Res. 2008, 78, 116–124. [Google Scholar] [CrossRef]
  258. Mandl, J.N.; Akondy, R.; Lawson, B.; Kozyr, N.; Staprans, S.I.; Ahmed, R.; Feinberg, M.B. Distinctive TLR7 signaling, type I IFN production, and attenuated innate and adaptive immune responses to yellow fever virus in a primate reservoir host. J. Immunol. 2011, 186, 6406–6416. [Google Scholar] [CrossRef] [PubMed]
  259. Lustig, S.; Olshevsky, U.; Ben-Nathan, D.; Lachmi, B.E.; Malkinson, M.; Kobiler, D.; Halevy, M. A live attenuated West Nile virus strain as a potential veterinary vaccine. Viral Immunol. 2000, 13, 401–410. [Google Scholar] [CrossRef] [PubMed]
  260. Wang, T.; Anderson, J.F.; Magnarelli, L.A.; Wong, S.J.; Koski, R.A.; Fikrig, E. Immunization of mice against West Nile virus with recombinant envelope protein. J. Immunol. 2001, 167, 5273–5277. [Google Scholar] [CrossRef] [PubMed]
  261. Xiao, S.Y.; Guzman, H.; Zhang, H.; Travassos da Rosa, A.P.; Tesh, R.B. West Nile virus infection in the golden hamster (Mesocricetus auratus): A model for West Nile encephalitis. Emerg. Infect. Dis. 2001, 7, 714–721. [Google Scholar] [CrossRef] [PubMed]
  262. Whitehorn, J.; Yacoub, S. Global warming and arboviral infections. Clin. Med. 2019, 19, 149–152. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A depiction of the virus life cycle and the inhibitor sites.
Figure 1. A depiction of the virus life cycle and the inhibitor sites.
Viruses 17 00074 g001
Figure 2. A depiction of the different approaches for developing antivirals against flavivirus.
Figure 2. A depiction of the different approaches for developing antivirals against flavivirus.
Viruses 17 00074 g002
Table 1. Antivirals targeting flavivirus entry.
Table 1. Antivirals targeting flavivirus entry.
TargetDrugViral SpecificityStudy StageRef.
Envelope Z2DENV, YFVIn vitro[5,6]
ZIKVIn vivo[5]
DN59DENV-2In vitro[7,8]
WNVIn vitro
P5JEV, ZIKVIn vivo[9]
DET2 and DET4DENV-2In vitro[10,11]
Dipeptide EFDENVIn vitro[12]
JBJ-01-162-04DENV, JEV, WNV, ZIKVIn vitro[13]
mAb513DENVIn vivo[14]
2D22DENVIn vivo[14,15,16]
ZIKV-IgZIKVPhase 1 clinical trial[17]
TY014YFVPhase 1 clinical trial[17]
TyzivumabZIKVPhase 1 clinical trial[17]
DN59DENV, WNVIn vitro[18]
MGAWN1WNVClinical trial withdrawn due to low enrollment[19]
3-110-22 (Cyanohydrazone)DENV, JEV, WNV, ZIKVIn vitro[20]
ZKA190 and FIT-1ZIKVIn vivo[21]
ZIKV-117ZIKVIn vivo[22]
EDE1 and EDE2ZIKVIn vitro[23]
mAb WNV-86WNVIn vivo[24]
GossypolDENV, ZIKVIn vitro[25]
CurcuminDENV, ZIKVIn vitro[26,27]
BP34610DNEV-1, 2, 3, 4, and JEVIn vitro[28]
Viral entryGeraniinDENV-2In vivo[29,30]
PalmatineWNV, DENV-2, JEV, YFV, ZIKVIn vitro[31]
Prochloroperazine (PCZ)DENV, JEVIn vitro[32]
DaptomycinJEVIn vitro[33,34,35]
Puerto Rico ZIKV
NanchangmycinCHIKV, DENV, WNV[34]
Erlotinib, SunitinibDENVIn vivo[36]
WNV, ZIKVIn vitro
25-HydroxylcholesterolDENV, YFV, WNVIn vitro[37]
ZIKVIn vivo
ChloroquineZIKVIn vivo[38,39]
DENVPhase 2 clinical trial failed (no viremia reduction)[33,40]
NiclosamideDENV, WNV, YFV, JEVIn vitro[41]
ZIKVIn vivo
DET4 and DET2DENVIn vitro[10,11]
EFDENVIn vitro[12]
Ri57DENV, ZIKVIn vitro[42]
AC-10ZIKVIn vivo[43]
Table 2. Host-directed flavivirus antivirals.
Table 2. Host-directed flavivirus antivirals.
TargetDrugViral SpecificityStudy StageRef.
Dopamine D2 receptorProchloroperazineDENV, JEVIn vitro[32]
ClathrinChlorpromazineJEV, WNVIn vitro[53]
AXLNanchangmycinZIKVIn vitro[34]
Lipoxygenase-activating proteinMK-591ZIKVIn vitro[35]
Receptor tyrosine kinase and epidermal growth factor tyrosine kinaseSunitinib and erlotinibDENVIn vivo[36]
Adenosine monophosphate-activated protein kinasePF-06409577, metformin, and AICARWNV, ZIKV and DENVIn vitro[54]
Holesterol-25-hydroxylase25-hydroxylcholesterolZIKVIn vitro[37]
Coenzyme acetylPF-05175157WNVIn vivo[55]
Sphingomyelinase-2GW4869WNV and ZIKVIn vitro[56,57]
Endosome acidificationChloroquineDENVIn vitro[58]
Endosomal acidificationNiclosamideZIKVIn vitro[59]
De novo pyrimidine biosynthesis pathwayAR-12, P12-23, and P12-34aDENV, ZIKV, and JEVIn vitro[60]
Table 3. Antivirals targeting flavivirus replication and polyprotein synthesis and processing.
Table 3. Antivirals targeting flavivirus replication and polyprotein synthesis and processing.
TargetDrugViral SpecificityStudy StageRef.
NS1Peptide 3, 4, 10, 11DENVIn vitro[68,69]
Deoxycalyxin-AZIKVIn silico[70]
mAb AA12ZIKVIn vivo[71]
mAb 2B7DENV, WNV, ZIKVIn vitro[72]
Castanospermine and deoxynojirimycinDENV, ZIKVIn vitro[73]
N-nonyl-DeoxynojirimycinDENV-2, JEVIn vitro[74]
CelgosivirDENVPhase 1 clinical trial[75,76]
NS2ANovobiocinDENV, ZIKVIn vivo[77,78]
NS2B–NS3TemoporfinDENV, YFV, WNV, JEVIn vitro[65]
ZIKVIn vivo[65]
JMX0207DENV-2In vitro[79]
ZIKVIn vivo[77]
NelfinavirDENV-2In vitro[80]
Compound 4ZIKVIn vitro[81]
Compound 14, Compound 15DENVIn vitro[82]
NSC135618DENV, ZIKV, WNV, YFVIn vitro[83]
ZP10ZIKVIn vitro[84]
MB21DENVIn vitro[85]
AprotininDENVIn vitro[85,86]
WNVIn silico
HydroxychloroquineZIKVIn vivo[87,88]
Methylene blueZIKVIn vivo[89]
DENVIn vitro[79,90,91]
Erythrosin BZIKV, DENVIn vitro
IvermectinYFV, WNVIn vitro[92]
ZIKVIn vivo[93]
DENVPhase 2/3 clinical trial[94]
Myricetin, quercetin, luteolin, isorhamnetin, apigenin, and curcuminZIKVIn vitro[51]
BortezomibZIKV, DENVIn vitro[95]
PalmatineDENV, JEV, WNV, ZIKVIn vitro[31]
Asunaprevir, SimeprevirZIKVIn vitro[96]
NS3ST-610DENVIn vivo[97]
SYC-1307DENV, JEV, WNV, ZIKVIn vivo[98]
ML283DENV, WNVIn vivo[99]
SuraminDENV, ZIKVIn vitro[100]
NS2BNSC157058WNVIn silico[101]
ZIKVIn vivo
NS4ACompound B and SBI-0090799DENV, ZIKVIn vitro[102]
NS3-NS4BJNJ-A07DENVIn vivo[103]
JNJ-64281802DENVPhase 2 clinical trial
JNJ-1802DENVPhase 1 clinical trial[104]
JEV, WNV, ZIKVIn vitro
NS4BNITD-688DENVIn vivo[105]
ManidipineJEVIn vivo[106]
DENV, ZIKV, WNVIn vitro
JMX0254 DENV, YFVIn vivo[107,108]
SBI-0090799ZIKVIn vitro[109]
AZD0530, dasatinibDENV-2In vitro[110,111]
NS5GalidesivirWNV, TBEV, ZIKVIn vivo[112,113]
YFVPhase I clinical trial[114]
FavipiravirWNV, YFVIn vitro[115]
ZIKVIn vivo[116]
BalapiravirDENVPhase 1/2 clinical trial[117]
NITD-008ZIKV, TBEV, DENVIn vivo[118,119]
AT-752DENV, YFVPhase II clinical trial[120,121]
WNV, ZIKV, JEVIn vitro
7DMATBEV, ZIKV, WNV, DENVIn vivo[111,122,123]
SofosbuvirYFVIn vivo[124]
ZIKVIn vivo[125,126]
NITD-434, NITD-640Pan-flavivirusIn vitro[127]
NITD-29DENVIn vitro[128]
Efavirenz, tipranavir, dasabuvirWNV, ZIKV, TBEVIn vitro[129]
AR-12DENVIn vivo[60]
P12-23, P12-34DENV, ZIKV, JEVIn vitro
RibavirinZIKVIn vivo[130]
IvermectinZIKVIn vitro[131]
DENVPhase 2/3 clinical trial[132]
T-1106YFVIn vivo[133]
RifapentineYFVIn vivo[6]
EmetineZIKVIn vivo[134]
LycorineZIKVIn vivo[135]
DolutegravirZIKVIn vitro[136]
Compound TPBZIKVIn vivo[137]
SinefunginWNV, DENVIn vivo[138,139,140]
ChartaceonesZIKVIn vitro[52]
FleximersDENV, ZIKV, YFVIn vitro[141,142]
Table 4. Antivirals targeting flavivirus assembly and egress.
Table 4. Antivirals targeting flavivirus assembly and egress.
TargetDrugViral SpecificityStudy StageRef.
AssemblyDeoxynojirimycin (DNJ) DENVIn vivo[74]
CastanospermineDENVIn vivo[187]
CelgosivirDENVPhase 1/2[188,189,199]
UV-4BDENVPhase 1/2[190]
UV-12DENVIn vivo[191,192,193]
CM-9-78
CM-10-18
PF-05175157WNV, DENV, ZIKVIn vivo[55]
TOFAIn vitro[55,194]
MEDICA 16In vitro
SFV785DENV, YFVIn vitro[195,196]
LovastatineDENVIn vivo[197,198,200]
Atorvastatine, ezetimibeDENV, ZIKVIn vitro[201]
CapsidVGTI-A3DENVIn vitro[194]
VGTI-A3-03In vitro[202]
ST-148In vivo[203,204]
Table 5. This table lists publications that implicate artificial/mi/si/sh RNAs as having potential against flaviviruses.
Table 5. This table lists publications that implicate artificial/mi/si/sh RNAs as having potential against flaviviruses.
Artificial/mi/si/sh RNAsViral SpecificityStudy StageRef.
amiRNA #1 and amiRNA #2JEVIn vitro[223]
amiRNA #1 and amiRNA #2WNVIn vitro[224]
amiRNA DENV-128DENVIn vitro[225]
miR-124aWNVIn vivo[228]
miR-155WNVIn vivo[229]
shRNAYFVIn vivo[230]
siRNAZIKVIn vivo[231]
Table 6. This table lists publications that implicate exosomes as having potential against flaviviruses.
Table 6. This table lists publications that implicate exosomes as having potential against flaviviruses.
Viral SpecificityStudy StageRef.
DENVIn vitro[237]
WNVIn vivo[238]
ZIKVIn vitro[239]
JEVIn vivo[240]
ZIKVIn vivo[241]
ZIKVIn vivo[242]
ZIKVIn vivo[243]
ZIKVIn vivo[244]
ZIKVIn vitro[241]
DENVIn vitro[245]
ZIKVIn vivo[246]
Table 7. Clinical trials on drugs targeting flaviviruses.
Table 7. Clinical trials on drugs targeting flaviviruses.
AntiviralTargetVirusClinical Trial IdentifierClinical TrialStatusNoteRef.
TY014EnvelopeYFVNCT03776786Phase 1CompletedSafe and abrogate viremiaClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Safety and Tolerability of an Antibody Against Yellow Fever Virus (TY014) in Humans/ClinicalTrials.gov/NCT03776786
Zika virus immune globulin (ZIKV-IG)EnvelopeZIKVNCT03624946Phase 1CompletedSafe and well toleratedClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Study in Healthy Volunteers Evaluating Safety and Pharmacokinetics of Zika Virus Immune Globulin (ZIKV-IG)/ClinicalTrials.gov/NCT03624946
Tyzivumab EnvelopeZIKVNCT03443830Phase 1CompletedNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Safety and Tolerability of an Antibody Against Zika Virus (Tyzivumab) in Humans/ClinicalTrials.gov/NCT03443830
Intravenous immunoglobulin (IVIG)EnvelopeJEVNCT01856205Phase 2CompletedJEV patients had a greater increase in neutralizing antibody titers Clinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Safety and Efficacy Study of Intravenous Immunoglobulin to Treat Japanese Encephalitis/ClinicalTrials.gov/NCT01856205
Omr-IgG-amEnvelopeWNVNCT00068055Phase 1/2CompletedSafe and well-tolerated in patientsClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/IVIG—West Nile Encephalitis: Safety and Efficacy/ClinicalTrials.gov/NCT00068055
MGAWN1Envelope WNVNCT00515385Phase 1CompletedSafe in healthy subjectsClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/A Trial to Evaluate the Safety of a Single Intravenous Infusion of MGAWN1 in Healthy Adults/ClinicalTrials.gov/NCT00515385
Carica papaya leaf extract (CPLE)NS2B–NS3DENVNCT06121934Phase 3CompletedNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Efficacy and Safety of Carica Papaya in Dengue Fever: A Randomised Clinical Trial//ClinicalTrials.gov/NCT06121934
Ribavirin NS3 helicaseJEVNCT00216268Phase 2UnknownNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Treatment of Japanese Encephalitis/ClinicalTrials.gov/NCT00216268
EYU688NS4BDENVNCT06006559Phase 2OngoingNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/A Study to Assess the Efficacy, Safety and Pharmacokinetics of EYU688 in Patients With Dengue Fever/ClinicalTrials.gov/NCT06006559
GalidesivirNS5YFVNCT03891420Phase 1TERMINATEDWithdrawn due to lack of fundingClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/A Study to Evaluate the Safety, Pharmacokinetics and Antiviral Effects of Galidesivir in Yellow Fever or COVID-19/ClinicalTrials.gov/NCT03891420
Iron-fortified foodOxidative stress-mediated signalingJEVNCT06027801Not ApplicableRecruitingNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Iron Fortified Food to Improve Japanese Encephalitis and Typhoid Fever Vaccine Immunogenicity/ClinicalTrials.gov/NCT06027801
MetforminAMPKYFVNCT04267809Phase 2CompletedNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Modulate Cellular Stress in the Immune Cells to Reduce Rate of Symptomatic Viral Infection/ClinicalTrials.gov/NCT04267809
AVI-4020 injectionInterfere with RNA–RNA duplex structuresWNVNCT00387283Phase 1CompletedNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Pharmacokinetic Study in Cerebral Spinal Fluid After a Single Dose of AVI-4020/ClinicalTrials.gov/NCT00387283
IvermectinIMPα/β (Host)DENVNCT02045069Phase 2/3UnknownNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Efficacy and Safety of Ivermectin Against Dengue Infection/ClinicalTrials.gov/NCT02045069
IvermectinIMPα/β (Host)DENVNCT03432442Phase 2CompletedSafe and accelerated NS1 antigen clearance in dengue patientsClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Pharmacokinetics and Pharmacodynamics of Ivermectin in Pediatric Dengue Patients/ClinicalTrials.gov/NCT03432442
UV-4BER α-glucosidase I and IIDENVNCT02061358Phase 1CompletedSafe in healthy subjectsClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Study to Determine the Safety, Tolerability and Pharmacokinetics of UV-4B Solution Administered Orally in Healthy Subjects/ClinicalTrials.gov/NCT02061358
ChloroquineEndosomal acidificationDENV, ZIKVNCT00849602Phase 1/2CompletedCQ ameliorated pain and lowered the viral load in dengue patientsClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/The Effect of Chloroquine in the Treatment of Patients With Dengue/ClinicalTrials.gov/NCT00849602
DexamethasoneNF-kB and activator factor-1DENVNCT05631405Not ApplicableOngoingNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Efficacy of Dengue Infection With Warning Signs Treated With Dexamethasone (DengDex Study)/ClinicalTrials.gov/NCT05631405
Anti-dFcγ receptor DENVNCT01443247Not ApplicableCompletedNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Role of Andi-d in Dengue Fever: a Pilot Study/ClinicalTrials.gov/NCT01443247
AnakinraInterleukin-1 receptor (IL-1R)DENVNCT05611710Phase 2OngoingNo results availableClinical trials.Gov (accessed on 1 July 2024); available online: Study Details/Anakinra in Dengue With Hyperinflammation (AnaDen)/ClinicalTrials.gov/NCT05611710
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tripathi, A.; Chauhan, S.; Khasa, R. A Comprehensive Review of the Development and Therapeutic Use of Antivirals in Flavivirus Infection. Viruses 2025, 17, 74. https://doi.org/10.3390/v17010074

AMA Style

Tripathi A, Chauhan S, Khasa R. A Comprehensive Review of the Development and Therapeutic Use of Antivirals in Flavivirus Infection. Viruses. 2025; 17(1):74. https://doi.org/10.3390/v17010074

Chicago/Turabian Style

Tripathi, Aarti, Shailendra Chauhan, and Renu Khasa. 2025. "A Comprehensive Review of the Development and Therapeutic Use of Antivirals in Flavivirus Infection" Viruses 17, no. 1: 74. https://doi.org/10.3390/v17010074

APA Style

Tripathi, A., Chauhan, S., & Khasa, R. (2025). A Comprehensive Review of the Development and Therapeutic Use of Antivirals in Flavivirus Infection. Viruses, 17(1), 74. https://doi.org/10.3390/v17010074

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop