key: cord-0908742-7aebgk0y authors: Sukmarini, Linda title: Antiviral Peptides (AVPs) of Marine Origin as Propitious Therapeutic Drug Candidates for the Treatment of Human Viruses date: 2022-04-19 journal: Molecules DOI: 10.3390/molecules27092619 sha: e9b33d1dbcedcb28ed4c6b7433ed6a229fd408ab doc_id: 908742 cord_uid: 7aebgk0y The marine environment presents a favorable avenue for potential therapeutic agents as a reservoir of new bioactive natural products. Due to their numerous potential pharmacological effects, marine-derived natural products—particularly marine peptides—have gained considerable attention. These peptides have shown a broad spectrum of biological functions, such as antimicrobial, antiviral, cytotoxic, immunomodulatory, and analgesic effects. The emergence of new virus strains and viral resistance leads to continuing efforts to develop more effective antiviral drugs. Interestingly, antimicrobial peptides (AMPs) that possess antiviral properties and are alternatively regarded as antiviral peptides (AVPs) demonstrate vast potential as alternative peptide-based drug candidates available for viral infection treatments. Hence, AVPs obtained from various marine organisms have been evaluated. This brief review features recent updates of marine-derived AVPs from 2011 to 2021. Moreover, the biosynthesis of this class of compounds and their possible mechanisms of action are also discussed. Selected peptides from various marine organisms possessing antiviral activities against important human viruses—such as human immunodeficiency viruses, herpes simplex viruses, influenza viruses, hepatitis C virus, and coronaviruses—are highlighted herein. Infectious diseases, mainly caused by viral pathogens, remain a primary global health issue that has already caused a high mortality rate [1] . Many of the most threatening or deadliest human infectious diseases are caused by viruses, such as human immunodeficiency viruses (HIVs) [2, 3] , influenza viruses [4, 5] , hepatitis viruses [3, 6] , and the recent emerging pandemic threat of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [7, 8] . In spite of striking development in vaccines and small-molecule antiviral drugs, new emerging and re-emerging viral disease outbreaks and the progression of antiviral drug resistance and drug toxicity have forced scientists to perpetually probe novel antiviral agents. Moreover, only a few therapeutic drugs are available for various viruses. Due to their excellent efficacy, high selectivity, and low potential for resistance development, the development of peptide-derived drugs to overcome their constraints on stability and bioavailability is becoming a point of interest in the pharmaceutical industry [9] [10] [11] . Hence, antiviral peptides (AVPs) that mainly originate from antimicrobial peptides (AMPs) with antiviral activities can be prospective antiviral agents to fight viral infections. AVPs are typically short peptides (generally consisting of 12-50 amino acid residues) with positively charged (typically +2 to +9) and amphipathic structures [12] [13] [14] [15] [16] [17] . In addition to these general features enabling these peptides to serve as antimicrobials (including antibacterials, antifungals, and antiparasitics), hydrophobicity is likely to be a key characteristic for AVPs to target enveloped viruses [18, 19] . It is worth mentioning that previous As specific antiviral drugs are mainly dedicated to treating particular viruses, different phases of the viral life cycle have been used to search for novel antiviral drugs. AVPs act against enveloped viruses by interrupting the fundamental stages of their life cycle of entry, synthesis, or assembly. Their inhibition sites include the viral particle or virion inhibition (virucidal effect), adsorption (cellular association), viral penetration, endosomal escape, viral uncoating, viral genome replication, and viral assembly, packaging, and release [14, 39] . Moreover, the proposed mechanism of action of antiviral activity of AVPs may generally encompass (1) their direct binding to the viral target, which is involved in the direct inhibition of host cell infection or viral pathogenesis; (2) their attachment to the target on the host surface (indirect inhibition), which is engaged in the competition or interaction with functional surface proteins of viruses; and (3) their indirect virus-targeting function through suppression of the viral gene expression, as well as inhibition of the viral enzymes, e.g., viral polymerase and integrase, related to the intracellular replication and transcription (biological function inhibition) ( Figure 1 ) [29, [39] [40] [41] [42] . The binding to host cells through interaction with functional receptors is the initial trend in viral infection. Once the viral particle attaches to a host cell, its genetic material is inserted into the cell during this initial attachment and penetration stage. Regarding direct virion inhibition, the AVP papuamide A has shown an immediate virucidal effect of HIV-1 inhibition through a viral-membrane-targeting mechanism [43, 44] . This marine peptide bears a tyrosine hydroxy and an aliphatic moiety tail that have been suggested to interact with the cholesterol membrane of the virus target and assist with insertion into the viral membrane, respectively, resulting in the subsequent viral membrane disruption and, ultimately, viral inactivation [44] . The indirect binding, as evidenced by the HIV-1-targeting α-defensin human neutrophil peptide HNP-1, has been found to bind viral envelope Env and host CD4 glycoproteins/ co-receptors in a glycan-and serum-independent manner. In addition, the oligomerization or refolding of HNP-1 could block the viral fusion. Hence, this peptide requires a multifaceted mode of action for HIV entry inhibition [45, 46] . Moreover, several studies [41, [47] [48] [49] [50] have reported that two mechanisms of the cellular association or viral attachment may prevent the entry of the enveloped viruses. In the instance of the activity towards the influenza virus, firstly, the AVP competes with sialic acid to bind with the envelope glycoprotein-namely, hemagglutinin (HA)-and clogs its receptor site, inhibiting the influenza virion from interacting with the host cell membrane. The peptide can imitate sialic acid's behavior to be recognized by the receptor-binding site of viral HA. Furthermore, the binding of AVP to the other main component of the influenza virus-namely, N-acetylneuraminic acid-on the host cell surface can also prevent viral attachment to host cells. Secondly, the conformation of HA is inhibited; thus, intracellular entry is prohibited, leading to endosomal escape and viral genome release [41] . A similar antiviral activity targeting primary attachment has also been found to inhibit HSV and hepatitis viruses. The AVP with a positive charge and good hydrophobicity binds to a cellular glycan moiety to prevent HSV from attaching to the host cell surface [51] . This glycan moiety is heparan sulfate, which is known to be a negatively charged glycosaminoglycan that favors HSV viral particles through its basic positively charged binding pocket in the virion glycoprotein to attach to the host cell surface. Therefore, the AVP can prevent HSV virion invasion by binding to glycosaminoglycan molecules as a receptor, hampering the interaction of the virus and the receptor in a host cell. With respect to hepatitis virusesparticularly HCV-some AVPs have also been demonstrated to interact with the virion receptors and co-receptors. AVPs that resemble the cellular protein apolipoprotein E (ApoE) can break up the glycan-dependent interaction or attachment of HCV, hindering entry and infection of target host cells (hepatocytes) [52, 53] . As observed by Chi et al. (2016) [54] , the HCV fusion-inhibitory peptide could block viral envelope glycoproteins' E1/E2-mediated membrane fusion by interfering with E1 and E2 heterodimerization. Moreover, the peptide was likely to provoke the dimer E1/E2 glycoproteins' conformational changes, impairing HCV membrane fusion. Additionally, another AVP (CL58) seemed to inhibit viral entry, possibly after initial binding (post-binding) of the co-receptor cellular membrane protein CD81, and just prior to the final intracellular fusion in endosomes [55, 56] . Furthermore, these mentioned inhibition mechanisms of the viral membrane fusion stage by peptides have been further comprehensively discussed elsewhere [57] . In addition, AVPs have also been reported to inhibit the viral replication of HSV-2, influenza viruses, and HCV. In the case of HSV-2, the AVPs can block the transport of a primary viral protein named VP16 into the nucleus. This transcriptional protein regulator induces immediate expression of viral genes required for survival at the initial cellular response. Some AVPs that act on influenza viruses also target viral-RNA-dependent RNA polymerase, comprising PB1, PB2, and PA subunits. These subunits control polymerization and endonuclease cleavage, recognition, binding to the host mRNAs, and endonuclease activity towards host pre-mRNA. Binding those subunits, AVPs can also prohibit the assembly of the influenza polymerase complex [13, 41, 58] . For HCV, AVPs can act on NS3-4A-a multifunctional enzyme with serine proteinase and helicase functions that are harnessed for HCV replication [59, 60] . Furthermore, to inhibit the replication of the very recent virus SARS-CoV-2, Tonk et al. (2021) [61] and Heydari et al. (2021) [62] gathered findings on a number of antiviral activities of AVPs that could be involved. Some AVPs directly act on the viral envelope (virucidal effect), binding to the viral spike glycoprotein that blocks the interaction with angiotensin-converting enzyme-2 (ACE2) in host cells, hampering endosomal acidification for uncoating throughout the initial viral life cycle, or escorting the host ACE2 receptor [63] [64] [65] [66] [67] . Moreover, several approaches have been deployed to evaluate the antiviral activity of marine peptides, including neutralization, viral titration, cell viability, and virus plaquereduction assays [27] . Some selected examples of marine AVPs that possess antiviral activity against important human enveloped viruses-such as HIV, influenza, HSV, HCV, and even SARS-CoV-2-are summarized in Figure 1 , listed in Table 1 , and featured below. Various commercially available anti-HIV drugs pose several restrictions, including toxicity, resistance, side effects, and long-term treatment, which have limited the application of these drugs. They may break down at a subsequent phase due to severe adverse effects and the emergence and spread of drug-resistant strains [25, 81] . However, the development of one of the marine-derived anti-HIV compounds entering clinical evaluation discloses the excellent potential of marine natural products, underlining the continuous prerequisite of progress in anti-HIV drugs derived from living entities. The compound is a lectin-based griffithsin originally obtained from a red alga [82, 83] . Many more potential anti-HIV therapeutics from marine resources have also been previously described [84, 85] . Moreover, recently reported AVPs derived from marine-sourced organisms have demonstrated anti-HIV activity, including mirabamides E-H, stellettapeptines A and B, mollamide F, malformin C, and divamide A ( Figure 2 and Table 1 ). Various commercially available anti-HIV drugs pose several restrictions, including toxicity, resistance, side effects, and long-term treatment, which have limited the application of these drugs. They may break down at a subsequent phase due to severe adverse effects and the emergence and spread of drug-resistant strains [25, 81] . However, the development of one of the marine-derived anti-HIV compounds entering clinical evaluation discloses the excellent potential of marine natural products, underlining the continuous prerequisite of progress in anti-HIV drugs derived from living entities. The compound is a lectin-based griffithsin originally obtained from a red alga [82, 83] . Many more potential anti-HIV therapeutics from marine resources have also been previously described [84, 85] . Moreover, recently reported AVPs derived from marine-sourced organisms have demonstrated anti-HIV activity, including mirabamides E-H, stellettapeptines A and B, mollamide F, malformin C, and divamide A ( Figure 2 and Table 1 ). The exploration of marine sponges as a prosperous source of antiviral lead structures [86] has led to the isolation of new Stelletta-derived mirabamides E-H ( Figure 2 ). Along with the known mirabamide C, these cyclic depsipeptides (cyclodepsipeptides) have been obtained from the sponge Stelletta clavosa Ridley collected from the Torres Strait, Queensland, Australia. In addition, the extraction of mirabamides from other phylogenetically different sponges associated with diverse microbes-such as Siliquariasspongia mirabilis-suggests that mirabamides are possibly derived from microbes or bacteria [68] . Structurally, the new cyclodepsipeptides mirabamides E-H are likely to be previously recognized peptide-based HIV-1 entry inhibitors-the mirabamides A-D and papuamides [87] . Moreover, based on a neutralization assay using pseudotyped viruses with an enveloped HIV-1 strain and tested with the known mirabamide C, these new cyclodepsipeptides exhibit considerable antiviral replication activity towards the genital epithelial cells expressing CD4 and CCR5 HIV-1 co-receptors (IC 50 = 121, 62, 68, and 41 nM, for mirabamides E-H, respectively). Hence, all four new depsipeptides seem to be potent inhibitors of HIV-1 at the entry stage of infection. Apparently, this class of mirabamides binds to the HIV-1 envelope glycoprotein to inhibit viral fusion into the host cell membrane [87] . However, mirabamides E-H differ in having an unprecedented 2-amino-2-butenoic acid moiety instead of threonine. It is noteworthy that the twofold elevation of the activity of mirabamides F and G compared with mirabamide C was likely due to the residue alteration. A similar change in the hydrophobicity of 2,3-dihydroxy-2,6,8-trimethyldeca-(4Z,6E)-dienoic acid (Dhtda) with 3-hydroxy-2,6,8-trimethyldeca-(4Z,6E)-dienoic acid (Htda) yielded a twofold increase in the inhibitory effect (observed with mirabamide E compared to mirabamide F, as well as with mirabamide G compared to mirabamide H). These results imply that the hydrophobicity is attributable to the potency of inhibition to some extent [68] . The other recent sponge genus Stelletta-derived anti-HIV cyclodepsipeptides-stellettapeptines A and B ( Figure 2 )-were successfully isolated from a Stelletta sp. obtained from northwestern Australia [69] . Stellettapeptines bear a 3-hydroxy-6,8-dimethylnon-4-(Z)-enoic acid-a unique non-canonical amino acid moiety of the polyketide subunit first identified in these cyclodepsipeptides. In addition, unprecedented 3-hydroxyglutamine and 3-hydroxyasparagine residues have also been found in these peptides. Similar to mirabamides, stellettapeptines are suggested to be derived from bacterial origin, as they possess the characteristic of non-ribosomal peptides or non-ribosomal peptide-polyketide hybrids, and are phylogenetically discrete from sponge-derived callipeptines and papuamides. Both of these new cyclic peptides were found to be remarkably active against HIV-1. Their antiviral activities have been assessed in an XTT-based cell viability assay employing HIV-1 RF -infected CEM-SS human T cells. Stellettapeptines A and B strongly inhibited HIV-1-infection-induced cytopathy, associated with apoptosis (EC 50 = 23 and 27 nM, respectively) [69] . The composition and cyclic configuration of non-canonical amino acids is likely of great importance for antiviral activity [88] . Therefore, similar to the cyclodepsipeptides mentioned above-papuamides and mirabamides-stellettapeptines may also target the viral membrane and act on viral entry. However, the further mechanism of viral entry inhibition of stellettapeptines has not been well described. In a conventional bioactivity-guided screening campaign of antiviral drug discovery targeting HIV by an international cooperative biodiversity group (ICBB) between the United States and Papua New Guinea, two new thiazoline cyclopeptides-mollamides E and F-have been discovered. Together with a new Tris-phenethyl urea called molleurea A, they were isolated from the extract of a tunicate Didemnum molle collected from Papua New Guinea [70] . It should be pointed out that those cyclopeptides were first recognized by metagenome sequencing of the symbiont cyanobacterium Prochloron didemni, whereby their predicted products were detected from a single colony of the ascidian, or by an Escherichia coli heterologous expression system of the engineered corresponding ribosomal biosynthetic route [89] . Nevertheless, Lu et al. (2012) [70, 90] reported that this finding constituted the first time these peptides had been fully depicted as isolated marine naturalproduct-based compounds. Moreover, the configuration of these peptides employing the advanced Marfey's approach showed that the thiazoline and phenylalanine in both mollamides E and F were in D-configuration instead of in the natural L-configuration of ribosomal peptides. Mollamide F showed HIV inhibition with an IC 50 value of 78 µM based on a cytoprotective assay, and an IC 50 value of 39 µM based on an HIV integrase inhibition assay, while no activity was observed in mollamide E in either assay. It has been previously reported that dolastatin 3, which is structurally similar to mollamides, exhibits HIV integrase activity [91] . HIV-1 integrase possessed by the virus is an enzyme vital for integrating the HIV genome or genetic material into the host genome within the cell, and is therefore regarded as an interesting target for therapeutic anti-HIV agents [92] . This would thus suggest that mollamide F could indirectly act on the viral target by inhibiting the biological function of the viral protein in terms of enzymatic activity of integrase. In addition to promising bioactive peptides from invertebrates and their bacterial symbionts as mentioned above, marine endophytic fungi have also been a source of propitious antiviral lead compounds [93, 94] . One of the more potent anti-HIV agents has been recently reported from a known mycotoxin compound-malformin C [71] . In addition to its original antibacterial activity [95, 96] , this fungal cyclic pentapeptide has also previously been found to bestow antiparasitic [97] and cytotoxic activities [98, 99] . Current advancements engaged in (meta)genomics and synthetic biology have also led to another breakthrough finding of excellent anti-HIV peptides from the genus Didemnum [72, 100] . Initially, the biological-activity-assay-guided fractionation of the tunicate D. molle E11-036 showed remarkable anti-HIV activity, unlike those from neighboring colony extracts of E11-037. The active compound was identified as divamide A ( Figure 2 )-a ribosomal lanthipeptide containing N-trimethylglutamate-and the partial precursor peptide sequence motif "GTTR." To further decipher structural confirmation, due to insufficient material, the compound was synthesized by the symbiotic cyanobacteria Prochloron didemni living in the marine tunicate Didemnum molle E11-036 through the expression of a div biosynthetic gene cluster. The gene cluster was reconstituted and expressed in Escherichia coli. Moreover, the generated semi-synthetic divamide A displayed significant HIV inhibition activity towards CEM TART -T cells, with an IC 50 value of 0.225 µM [72] . Likewise, other lanthipeptides, such as cinnamycin and duramycin, show high binding affinity and specificity to phosphatidylethanolamine (PE); divamide A presumably interacts with this viral envelope lipid, thereby blocking viral entry [101] . PE is abundant in enveloped viruses, including HIV-1, in which it is derived from the budding process at the plasma membrane of infected cells. [102] [103] [104] [105] . Due to its extensive distribution in many enveloped viruses, viral lipid PE-targeted AVPs such as lanthipeptides possess a wide spectrum of activity against various human pathogenic viruses, including Ebola, Dengue, West Nile, and Zika viruses-exemplified by duramycins [106, 107] -and HSV-1, as presented by cinnamycin [107] [108] [109] . One of the emerging and drug-resistant infectious diseases that has received significant attention is pneumonia or respiratory infections caused by influenza viruses. The most notable feature of influenza viruses is their expeditious evolution, generating exten-sive variability of strains-particularly the influenza A virus. There have been several outbreaks of influenza viruses, including H1N1, or Spanish flu (1918); H2N2, or Asian flu (1957); H3N2, or Hong Kong flu (1968); and the recent generation of influenza viruses in South-East Asia (A/H5N1; 1998) and the Netherlands and Belgium (A/H7N7; 2003). The many antiviral drugs used for influenza treatment include neuraminidase inhibitors (e.g., zanamivir and oseltamivir) as well as adamantane drugs (e.g., rimantadine and amantadine), which are not able to minimize the risk of adverse effects [25] . Moreover, one of the recent promising AVPs with inhibitory effects against influenza viruses but less toxicity is marine-fungus-derived asperterrestide A [73] (Figure 3 and Table 1 ). One of the emerging and drug-resistant infectious diseases that has received significant attention is pneumonia or respiratory infections caused by influenza viruses. The most notable feature of influenza viruses is their expeditious evolution, generating extensive variability of strains-particularly the influenza A virus. There have been several outbreaks of influenza viruses, including H1N1, or Spanish flu (1918); H2N2, or Asian flu (1957); H3N2, or Hong Kong flu (1968); and the recent generation of influenza viruses in South-East Asia (A/H5N1; 1998) and the Netherlands and Belgium (A/H7N7; 2003). The many antiviral drugs used for influenza treatment include neuraminidase inhibitors (e.g., zanamivir and oseltamivir) as well as adamantane drugs (e.g., rimantadine and amantadine), which are not able to minimize the risk of adverse effects [25] . Moreover, one of the recent promising AVPs with inhibitory effects against influenza viruses but less toxicity is marine-fungus-derived asperterrestide A [73] (Figure 3 and Table 1 ). Asperterrestide A-a cyclotetrapeptide with a rare 3-hydroxy-N-methyl-Phe moiety-was obtained from the cultivation broth of the marine gorgonian Echinogorgia aurantiaca-derived fungus A. terreus SCSGAF1062. In addition to its cytotoxicity against cancer cells, asperterrestide A has demonstrated inhibitory effects on an M2-resistant influenza virus strain A/W/33 (H1N1) and an M2-sensitive strain A/Hong Kong/8/68 (H3N2), with IC50 values of 20.2 and 0.41 μM, respectively, based on a cytopathic assay. It is likely that the potential of its antiviral activity was due to the presence of the rare moiety mentioned above [73] . However, this is a rather insufficient understanding of the antiviral mode of action of this fungal-derived AVP. The first marine-derived compound with antiviral activity that reached the market was vidarabine (Ara A). This sponge-derived nucleoside compound was licensed and approved as an anti-HSV drug [86] . Since it was later found to be less efficient than the newer commercial drug Zovirax (acyclovir), vidarabine was discontinued. However, due to viral resistance to acyclovir, more effective new antiviral drugs with high selectivity remain to be developed-especially those from peptide-based marine AVPs. These include marine fungal peptides, marine invertebrate peptides, and marine vertebrate peptides, which have all been found to demonstrate anti-HSV activities ( Figure 4 and Table 1 ). Asperterrestide A-a cyclotetrapeptide with a rare 3-hydroxy-N-methyl-Phe moietywas obtained from the cultivation broth of the marine gorgonian Echinogorgia aurantiacaderived fungus A. terreus SCSGAF1062. In addition to its cytotoxicity against cancer cells, asperterrestide A has demonstrated inhibitory effects on an M2-resistant influenza virus strain A/W/33 (H1N1) and an M2-sensitive strain A/Hong Kong/8/68 (H3N2), with IC 50 values of 20.2 and 0.41 µM, respectively, based on a cytopathic assay. It is likely that the potential of its antiviral activity was due to the presence of the rare moiety mentioned above [73] . However, this is a rather insufficient understanding of the antiviral mode of action of this fungal-derived AVP. The first marine-derived compound with antiviral activity that reached the market was vidarabine (Ara A). This sponge-derived nucleoside compound was licensed and approved as an anti-HSV drug [86] . Since it was later found to be less efficient than the newer commercial drug Zovirax (acyclovir), vidarabine was discontinued. However, due to viral resistance to acyclovir, more effective new antiviral drugs with high selectivity remain to be developed-especially those from peptide-based marine AVPs. These include marine fungal peptides, marine invertebrate peptides, and marine vertebrate peptides, which have all been found to demonstrate anti-HSV activities ( Figure 4 and Table 1 ). [110, 111] , based on the amino acid sequence from GenBank with accession number AEZ79080.1 [112] . Moreover, the synthetic Pa-MAP is derived from the antifreeze peptide (AFP) HPLC-8 b (as shown by the amino acid sequence), the structural model of which was retrieved from the SWISS-MODEL repository (https://swissmodel.expasy.org/repository, accessed [110, 111] , based on the amino acid sequence from GenBank with accession number AEZ79080.1 [112] . Moreover, the synthetic Pa-MAP is derived from the antifreeze peptide (AFP) HPLC-8 b (as shown by the amino acid sequence), the structural model of which was retrieved from the SWISS-MODEL repository (https://swissmodel.expasy.org/repository, accessed on 23 February 2022) [113] , with the UniProtKB AC number Q99013 (ANPB_ PSEAM) [114] . The blue and red colors within the structural model indicate positively and negatively charged residues, respectively. In addition to the potent anti-HIV peptide malformin C and anti-influenza peptide asperterrestide A, a number of recently isolated and reported marine-derived fungal peptides exerting anti-HSV properties are represented by aspergillipeptides D and E [74] , simplicilliumtide J, verlamelines A and B [75] , and acremonpeptides A and B, as well as Al(III)-acremonpeptide D [76] (Figure 4) . Aspergillipeptides D and E were isolated from a fermentation broth of the marine-gorgonian-derived fungus Aspergillus sp. SCSIO 41501. The fungus was isolated from the gorgonian Melitodes squamata collected from the South China Sea. Aspergillipeptide D is a new cyclopentapeptide, while its counterpart aspergillipeptide E is a new linear peptide which was likely the direct product of a high concentration of L-tryptophan added to the medium. Based on a plaque-reduction assay, aspergillipeptides D and E had significant antiviral activity against HSV-1 (IC 50 = 9.5 µM and 19.8 µM, respectively). Moreover, aspergillipeptide D also showed a remarkable antiviral effect against acyclovir-resistant clinical (ACV) isolates of HSV-1 (HSV-1-106 and HSV-1-135), with an IC 50 value of about 12.5 µM [74] . Further study of its mechanism of antiviral action revealed that this cyclic pentapeptide seemed not to act on viral particle inactivation, attachment, or penetration in the early stages of infection, but rather on the inhibition of the viral glycoprotein gB. As the most highly conserved glycoprotein and a class III viral fusion protein, gB engages in the interaction and fusion of the virion and the host cell membrane in a direct manner. The study showed that aspergillipeptide D decreased the expression and synthesis of the gB1 protein and its localization in the endoplasmic and Golgi apparatus, leading to reduced viral intercellular spread. Moreover, in the presence of the peptide, the number of cellular proteins related to translation, ribosomal structure, biogenesis, posttranslational modification, protein turnover, chaperones, and the cytoskeleton-which interact with gB-were also extensively decreased. Therefore, these results highlight the great potential of aspergillipeptide D for HSV-1 infection therapy-particularly for ACV-resistant strains [115] -which is in accordance with the computational-peptidology-approach-based study by Glossman-Mitnik et al. [116] . Their very recent computational analysis of the chemical reactivity and bioactive properties corroborated the potential of this AVP through its interaction with different target receptors. Indeed, the in silico ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) characteristics of aspergillipeptide D showed the absence of toxicity as well as fine absorption and distribution properties [116] . Regarding simplicilliumtide J, this new cyclohexadepsipeptide, along with previously known analogues verlamelines A and B, has been isolated from the marine fungal strain Simplicillium obclavatum EIODSF 0210 collected from marine sediment in the depths of the East Indian Ocean. Simplicilliumtide J had a notable anti-HSV-1 activity, with an IC 50 value of 14 µM, while the known analogues verlamelines A and B were first reported to possess antiviral activity against HSV-1, with IC 50 values of 16.7 and 15.6 µM, respectively, based on a plaque assay. Based on the structure-activity relationship analysis, their potent antiviral activity could be attributed to the presence of the lactonized 5-hydroxytetradecanoic acid moiety in these cyclic hexapeptides [75] . However, the mechanism of antiviral activity of these cyclopeptides remains puzzling. Additionally, with regard to marine fungal anti-HSV peptides, Luo et al. (2019) [76] also reported new antiviral cyclopeptide siderophores bearing hydroxamate isolated from the marine fungus Acremonium persicinum SCSIO 115, namely, acremonpeptides A and B, and the aluminum complex of acremonpeptide D, or Al(III)-acremonpeptide D. Interestingly, these cyclopeptides are defined by three 2-amino-5-(N-hydroxyacetamido)pentanoic acid (N 5 -hydroxy-N 5 -acetyl-L-ornithine) metal-ion-chelating moieties. They found that these peptides possessed antiviral activity against HSV-1, with EC 50 values of 16, 8.7 , and 14 µM, respectively. A further mode-of-action study revealed that these AVPs could inhibit viral DNA replication, as shown by the downregulated UL42 gene transcription in HSV. In addition to sponges and tunicates as the richest sources of antiviral compounds from marine invertebrates [86, 100, 117, 118] , several compounds from mollusks have also been found to have antiviral activity, such as anti-HSV activity. One recent example of marine-mollusk-derived AVPs is myticin C. Myticin C was found in mussel hemolymph and hemocytes of the Mediterranean mussel Mytilus galloprovincialis. This cysteine-rich antimicrobial peptide initially showed antiviral activity against an aquatic pathogen fish rhabdovirus-ostreid herpesvirus 1 (OsHV-1) [119] . The antiviral activity of this peptide against OsHV-1 led to further investigation of antiviral effects against human HSVs, including HSV-1 and HSV-2. Interestingly, a modified synthetic myticin C with a Tat-derived cell-penetrating motif (Myt-Tat) and the nanoencapsulation peptide (with nonionic surfactant nanovesicles or nanosomes) showed antiviral inhibition of HSV-1 and HSV-2, as indicated by their SI values. The putative SI values for Myt-tat were >8.21 towards HSV-1 and >10.5 towards HSV-2, whereas for encapsulated myticin C they were >7.69 towards HSV-1 and >8.32 towards HSV-2. In addition, based on the virus plaque titers, the encapsulated peptides exerted a viral-inhibitory effect at 22.5 µM or above, implying thorough suppression of viral replication. The nanovesicles can fuse with the plasma membrane, delivering the peptide into the cytoplasm. The study suggested that myticin peptides selectively block an intracellular stage of the viral replication course, rather than direct viral inactivation or a virucidal effect. However, further investigation of the detailed mode of antiviral action of these peptides is still required [77] . Moreover, concerning marine-vertebrate-derived AVPs, the promiscuous peptide Pa-MAP (multiple active peptides from Pleuronectes americanus) was derived from the antifreeze peptide (AFP) HPLC-8, originally isolated from the skin of the polar fish P. americanus [120] . This synthetic alanine-rich peptide has shown antiviral activity against the viruses HSV-1 and HSV-2. Based on a titer-reduction assay, the peptide caused 82% HSV-1 inhibition (at 45 µM) and 90% HSV-2 inhibition (at 23 µM), while 94% and 97% inhibition of HSV-1 and HSV-2, respectively, were observed at the concentration of 90 µM. Thus, these results suggest that the hydrophobic residue in this peptide could facilitate interaction with the viral envelope and phospholipids at the viral surface for inhibiting viral infection [121] . Moreover, further investigation by Franco et al. [78] proved that Pa-MAP exerts a direct virucidal effect, preventing viral adsorption or entry, which supports the previous finding [121, 122] . Viral hepatitis C, which leads to cirrhosis, carcinoma, and liver failure in humans, continues to be a serious global health burden. However, there is still no protective vaccine available for this disease. Moreover, despite its significant advancements, HCV therapy still faces the major issues of high cost and adverse effects. Thus, there is an urgent need to discover and develop potent anti-HCV drugs, including those derived from marine natural products. In recent years, some marine-microbial-derived peptides have been reported to have promising anti-HCV properties, including cyclo(L-Tyr-L-Pro) diketopiperazine [79] , valinomycin. and its analogues streptodepsipeptide P11A and streptodepsipeptide SV21 [80] (Figure 5 and Table 1) . Cyclo(L-Tyr-L-Pro) is a cyclic dipeptide diketopiperazine isolated from the Red Sea black sponge Spongia officinalis-associated fungus Aspergillus versicolor. This cyclodipeptide diketopiperazine exhibited potent activity against HCV NS3-4A protease, with an IC 50 value of 8.2 µg mL −1 [79] . This would thus suggest that this compound targets viral replication. Cyclo(L-Tyr-L-Pro) is a cyclic dipeptide diketopiperazine isolated from the Red Sea black sponge Spongia officinalis-associated fungus Aspergillus versicolor. This cyclodipeptide diketopiperazine exhibited potent activity against HCV NS3-4A protease, with an IC50 value of 8.2 μg mL −1 [79] . This would thus suggest that this compound targets viral replication. Along with the known valinomycin and its analogue streptodepsipeptide P11A, another new analogue-cyclodepsipeptide streptodepsipeptide SV21-was very recently isolated from a sea-cucumber-associated bacterium Streptomyces sp. SV21, collected from Along with the known valinomycin and its analogue streptodepsipeptide P11A, another new analogue-cyclodepsipeptide streptodepsipeptide SV21-was very recently isolated from a sea-cucumber-associated bacterium Streptomyces sp. SV21, collected from Lampung, Indonesia [80] . Indeed, the antiviral activity of valinomycin has been recently reported to inhibit the coronavirus SARS-CoV-2 [123] ; however, these current reported antivirals valinomycin and its derivatives demonstrated very significant inhibition of infectivity against HCV compared with the positive control epigallocatechin gallate (about 0-5% infectivity vs. 20% infectivity) [80] . Epigallocatechin gallate has been regarded as a strong HCV inhibitor, blocking early viral entry and attachment to the cell surface [124] . However, the mode of anti-HCV activity of these compounds has not been evaluated, although it has been speculated that valinomycin interferes with the binding of the coronavirus glycoprotein to the host cells [125] . However, it has been suggested that the symmetry of the ring system is likely vital for the bioactivity of valinomycin and those analogues, based on the structure-activity relationship analysis [80] . Several literature reviews have been extensively compiled in an attempt to search for potential antimicrobial peptides against SARS-CoV-2 [28, 61, 62, 67, 126, 127] . Moreover, drug repurposing to accelerate the identification of available drugs that can be used to treat coronavirus disease 2019 (COVID- 19) has also been broadly discussed [128] [129] [130] [131] [132] [133] [134] [135] . However, a recent marine AVP with potential anti-SARS-CoV-2 activity is exemplified by plitidepsin ( Figure 6 and Table 1 ), which is known under the trademark Aplidin ® (PharmaMar, S.A., Madrid, Spain) for the treatment of multiple myeloma [136] [137] [138] . Lampung, Indonesia [80] . Indeed, the antiviral activity of valinomycin has been recently reported to inhibit the coronavirus SARS-CoV-2 [123] ; however, these current reported antivirals valinomycin and its derivatives demonstrated very significant inhibition of infectivity against HCV compared with the positive control epigallocatechin gallate (about 0-5% infectivity vs. 20% infectivity) [80] . Epigallocatechin gallate has been regarded as a strong HCV inhibitor, blocking early viral entry and attachment to the cell surface [124] . However, the mode of anti-HCV activity of these compounds has not been evaluated, although it has been speculated that valinomycin interferes with the binding of the coronavirus glycoprotein to the host cells [125] . However, it has been suggested that the symmetry of the ring system is likely vital for the bioactivity of valinomycin and those analogues, based on the structure-activity relationship analysis [80] . Several literature reviews have been extensively compiled in an attempt to search for potential antimicrobial peptides against SARS-CoV-2 [28, 61, 62, 67, 126, 127] . Moreover, drug repurposing to accelerate the identification of available drugs that can be used to treat coronavirus disease 2019 (COVID- 19) has also been broadly discussed [128] [129] [130] [131] [132] [133] [134] [135] . However, a recent marine AVP with potential anti-SARS-CoV-2 activity is exemplified by plitidepsin ( Figure 6 and Table 1 ), which is known under the trademark Aplidin ® (PharmaMar, S.A., Madrid, Spain) for the treatment of multiple myeloma [136] [137] [138] . Plitidepsin is a cyclodepsipeptide originally isolated from the Mediterranean marine tunicate Aplidium albicans. This cyclodepsipeptide can now be obtained via total synthesis [139] . Computationally aided in silico studies of plitidepsin by molecular docking and molecular dynamic simulation related to its protease-inhibitory activity have shown its great binding effectiveness with the main protease of SARS-CoV-2 compared with other available drugs, e.g., hydroxychloroquine, remdesivir, and lopinavir. Thus, this peptide has shown tremendous potential for drug repurposing against COVID-19 [140, 141] . Moreover, very recently, White et al. (2021) [142] verified that plitidepsin exhibited anti-SARS-CoV-2 activity, with an IC90 value of 0.88 nM, which was stronger than remdesivir in the human cell line hACE2-293T by a factor of 27.5, with limited toxicity. By employing a drug-resistant mutant (a mutated eukaryotic translation elongation factor 1A or eEF1A in 293T cells), they also demonstrated that the antiviral activity of plitidepsin was assisted by the prohibition of the known target eEF1A, which is a vital host factor for viral replication. In addition, it has been demonstrated that plitidepsin treatment could reduce the replication of SARS-CoV-2 by two orders of magnitude and diminish lung inflammation in vivo in murine models. Hence, these gathered findings show that Plitidepsin is a cyclodepsipeptide originally isolated from the Mediterranean marine tunicate Aplidium albicans. This cyclodepsipeptide can now be obtained via total synthesis [139] . Computationally aided in silico studies of plitidepsin by molecular docking and molecular dynamic simulation related to its protease-inhibitory activity have shown its great binding effectiveness with the main protease of SARS-CoV-2 compared with other available drugs, e.g., hydroxychloroquine, remdesivir, and lopinavir. Thus, this peptide has shown tremendous potential for drug repurposing against COVID-19 [140, 141] . Moreover, very recently, White et al. (2021) [142] verified that plitidepsin exhibited anti-SARS-CoV-2 activity, with an IC 90 value of 0.88 nM, which was stronger than remdesivir in the human cell line hACE2-293T by a factor of 27.5, with limited toxicity. By employing a drug-resistant mutant (a mutated eukaryotic translation elongation factor 1A or eEF1A in 293T cells), they also demonstrated that the antiviral activity of plitidepsin was assisted by the prohibition of the known target eEF1A, which is a vital host factor for viral replication. In addition, it has been demonstrated that plitidepsin treatment could reduce the replication of SARS-CoV-2 by two orders of magnitude and diminish lung inflammation in vivo in murine models. Hence, these gathered findings show that plitidepsin is a promising therapeutic candidate for COVID-19. In addition to successful assessment through a phase I/II clinical trial by PharmaMar, it is now undergoing phase II/III trials for the treatment of COVID-19 [137, 142] . Results from in vitro investigations also demonstrate that the peptide has a potent antiviral function against all variants, with viral load reduction of 99% in animal lung tissue. Moreover, positive effects in phase I and II clinical trials on patients hospitalized with COVID-19 were also recorded [143] . In addition, Guisado-Vasco et al. have also recently reported that plitidepsin succeeded in providing the cure for prolonged viral SARS-CoV-2 replication in a patient with hematological malignancy and depleted B cells [144] . In summary, marine organisms such as sponges, tunicates, mollusks, microbes, and fish have recently produced potent peptides with moderate-to-significant antiviral activity in vitro. AVPs obtained or reported from these marine sources during the period covered in this short review, from 2011 to 2021, comprise about 25 compounds. Most of them are fungal-sourced natural peptides, and are mainly derived from non-ribosomal peptides. Conversely, ribosomal divamide A, myticin C, and Pa-MAP are (semi-)synthetic variants whose origins are amenable for structural modifications. These recently discovered marine cationic peptides act against the enveloped viruses via different mechanisms. Some AVPs have been known to be virucidal, act on viral entry, bind to the viral envelope, or inhibit viral replication and intercellular spread, while the antiviral activity of some others remains elusive. Interestingly, it has been markedly shown that almost all of these recently reported promising peptides are cyclic peptides, ranging from cyclic dipeptide diketopiperazine to cyclodepsipeptides. Due to its reduced conformational flexibility, the cyclic feature has been suggested to facilitate increased cell permeability and better bioactivity compared to those with a linear structure. Indeed, the cyclopeptide aspergillipeptide D was found to be more active than the linear peptide aspergillipeptide E. Cyclopeptides are also more resistant to degradative proteases due to their lack of C-and N-termini, and are endowed with entropic (Gibbs free energy) benefits in terms of molecular binding and recognition [145] [146] [147] . Therefore, these favorable properties make this class of peptides a captivating candidate for therapeutics. Improved therapeutic targeting can be attained through structural changes, including cyclization, the use of stable D-amino acids, and chemical modification. However, critical issues remain in the development of cyclopeptides for therapeutics, including bioavailability and cell permeability. Innovative methods could overcome these challenges, such as the application of cell-penetrating peptides and nanoencapsulation [146] [147] [148] [149] , as applied in the antiviral peptide myticin C (myt-Tat) against HSV-1 and HSV-2 [77] . Although several marine peptides are currently being subjected to preclinical and clinical investigations, or have even hit the market [22] , none of them are antiviral lead compounds or drugs yet. In vivo animal evaluation or therapeutic application have not yet been reported for almost all of the aforementioned promising AMPs with antiviral activity. However, through drug repurposing to cope with COVID-19, the synthetic anticancer drug plitidepsin has demonstrated anti-SARS-CoV-2 effects in vitro and in vivo, and is now undergoing phase II/III clinical trials. With the advancement in (meta)genomic and synthetic biology approaches, marine-peptide-based antiviral drug discovery and development will be accelerated. Fermentation technology-especially for microbial-metabolite-based peptides-can replace chemical synthesis for their scalable production. Furthermore, given their promising antiviral activities, the potential of AVPs with possibly reduced toxicity and fewer adverse effects in preclinical and clinical treatments should be further explored, aided by virtual screening via computational peptidology [150] . Apparently, marine peptides will play a pivotal role in developing lead compounds and drugs for various viral infectious diseases in the near future. Emerging and reemerging infectious diseases: Global overview Accountability for the Global Health Sector Strategies 2016-2021: Actions for Impact Estimates of global seasonal influenza-associated respiratory mortality: A modelling study Influenza virus-related critical illness: Prevention, diagnosis, treatment Viral hepatitis: Etiology, epidemiology, transmission, diagnostics, treatment, and prevention SARS-CoV-2 pandemic: An overview Emerging pandemic diseases: How we got to COVID-19 Current scenario of peptide-based drugs: The key roles of cationic antitumor and antiviral peptides Peptides, peptidomimetics, and polypeptides from marine sources: A wealth of natural sources for pharmaceutical applications Antimicrobial peptides as therapeutic agents: Opportunities and challenges Antimicrobial peptides: An emerging category of therapeutic agents Antimicrobial peptides: Features, action, and their resistance mechanisms in bacteria Antiviral peptides as promising therapeutic drugs From life in the sea to the clinic: The marine drugs approved and under clinical trial Antibiofilm activity of host defence peptides: Complexity provides opportunities Chemically modified and conjugated antimicrobial peptides against superbugs Peptide entry inhibitors of enveloped viruses: The importance of interfacial hydrophobicity Large-scale analysis of antimicrobial activities in relation to amphipathicity and charge reveals novel characterization of antimicrobial peptides APD: The antimicrobial peptide database The importance of amino acid composition in natural amps: An evolutional, structural, and functional perspective Marine organisms as a rich source of biologically active peptides. Front Marine peptides: Structure, bioactivities, and a new hope for therapeutic application Antifungal and antiviral products of marine organisms Role of marine natural products in the genesis of antiviral agents Marine peptides and their anti-infective activities Ten-year research update review: Antiviral activities from marine organisms Microbial metabolites: The emerging hotspot of antiviral compounds as potential candidates to avert viral pandemic alike COVID-19 Peptide-based antiviral drugs Antiviral peptides: Identification and validation AVPdb: A database of experimentally validated antiviral peptides targeting medically important viruses AntiVPP 1.0: A portable tool for prediction of antiviral peptides Antiviral peptides as promising therapeutics against SARS-CoV-2 Ribosomal peptide natural products: Bridging the ribosomal and nonribosomal worlds Bioactive peptide natural products as lead structures for medicinal use Ribosomally synthesized and post-translationally modified peptide natural products: Overview and recommendations for a universal nomenclature Diversity of nature's assembly lines-recent discoveries in non-ribosomal peptide synthesis Nonribosomal peptide synthesis-Principles and prospects Current progress in antiviral strategies A review: Mechanism of action of antiviral drugs Perspective of use of antiviral peptides against influenza virus Antiviral peptides with in vivo activity: Development and modes of action Papuamides A-D, HIV-inhibitory and cytotoxic depsipeptides from the sponges Theonella mirabilis and Thenonella swinhoei collected in Papua New Guinea Characterizing the anti-HIV activity of papuamide A Multifaceted mechanisms of HIV-1 entry inhibition by human α-defensin Defensins : A double-edged sword in host immunity Inhibition of influenza virus infections by sialylgalactose-binding peptides selected from a phage library mSialic acid-mimic peptides as hemagglutinin inhibitors for anti-influenza therapy A novel family of peptides with potent activity against influenza a viruses A new class of synthetic antilipopolysaccharide peptides inhibits influenza A virus replication by blocking cellular attachment Anti-HSV activity of lactoferricin analogues is only partly related to their affinity for heparan sulfate Hepatitis C virus attachment mediated by apolipoprotein E binding to cell surface heparan sulfate Hepatitis C virus glycan-dependent interactions and the potential for novel preventative strategies Identification of a potent and broad-spectrum hepatitis C virus fusion inhibitory peptide from the E2 stem domain A human claudin-1-derived peptide inhibits hepatitis C virus entry Entry inhibitors: New advances in HCV treatment Inhibition of viral membrane fusion by peptides and approaches to peptide design Disruption of the viral polymerase complex assembly as a novel approach to attenuate influenza A virus Hepatitis C viral NS3-4A protease activity is enhanced by the NS3 helicase Hepatitis C virus NS3/4A protease inhibitors: A light at the end of the tunnel Compelling evidence for the activity of antiviral peptides against SARS-CoV-2. Viruses Antiviral peptides against Coronaviridae family: A review Brilacidin demonstrates inhibition of SARS-CoV-2 in cell culture A broad-spectrum virusand host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2 Human intestinal defensin 5 inhibits SARS-CoV-2 invasion by cloaking ACE2 The potential of antiviral peptides as COVID-19 therapeutics Mirabamides E-H, HIVinhibitory depsipeptides from the sponge Stelletta clavosa Stellettapeptins A and B, HIV-inhibitory cyclic depsipeptides from the marine sponge Stelletta sp Thiazoline peptides and a tris-phenethyl urea from Didemnum molle with anti-HIV activity Aspernigrins with anti-HIV-1 activities from the marine-derived fungus Aspergillus niger SCSIO Jcsw6F30 Accessing chemical diversity from the uncultivated symbionts of small marine animals Asperterrestide A, a cytotoxic cyclic tetrapeptide from the marine-derived fungus Aspergillus terreus SCSGAF0162 Antiviral peptides from marine gorgonian-derived fungus Aspergillus sp. SCSIO 41501 Antifungal and antiviral cyclic peptides from the deep-sea-derived fungus Simplicillium obclavatum EIODSF 020 Natural hydroxamate-containing siderophore acremonpeptides A-D and an aluminum complex of acremonpeptide D from the marine-derived Acremonium persicinum SCSIO 115 Antiviral activity of myticin C peptide from mussel: An ancient defense against herpesviruses Linear antimicrobial peptides with activity against herpes simplex virus 1 and aichi virus Anti-HCV protease of diketopiperazines produced by the Red Sea sponge-associated fungus Aspergillus versicolor Anti-Infective and antiviral activity of valinomycin and its analogues from a sea cucumber-associated bacterium, Streptomyces sp. SV 21 Recent advances in anti-HIV natural products Impact of Q-Griffithsin anti-HIV microbicide gel in non-human primates: In situ analyses of epithelial and immune cell markers in rectal mucosa Griffithsin, a highly potent broad-spectrum antiviral lectin from red algae: From discovery to clinical application Potential anti-HIV agents from marine resources: An overview Marine natural products with anti-HIV in the last decade Antiviral lead compounds from marine sponges Mirabamides A-D, depsipeptides from the sponge Siliquariaspongia mirabilis that inhibit HIV-1 fusion Mutremdamide A and koshikamides C-H, peptide inhibitors of HIV-1 entry from different theonella species Variation in tropical reef symbiont metagenomes defined by secondary metabolism Ribosomal route to small-molecule diversity Dolastatin 3 and two novel cyclic peptides from a Palauan collection of Lyngbya majuscula A comprehensive review of bioactive peptides from marine fungi and their biological significance Structural diversity and biological activities of cyclic depsipeptides from fungi Malformin C, a new metabolite of Aspergillus niger Production and antibacterial activity of malformin C, a toxic metabolite of Aspergillus niger Solid-phase synthesis and biological activity of malformin C and its derivatives Fungal malformins inhibit bleomycin-induced G2 checkpoint in Jurkat cells Study of malformin C, a fungal source cyclic pentapeptide, as an anti-cancer drug Secondary metabolites of the genus didemnum: A comprehensive review of chemical diversity and pharmacological properties Antiviral activities and applications of ribosomally synthesized and post-translationally modified peptides (RiPPs) Lantibiotics as probes for phosphatidylethanolamine Functional organization of the HIV lipid envelope HIV-1 Assembly, Budding, and Maturation. Cold Spring Harb Role of lipids in virus replication Virion-associated phosphatidylethanolamine promotes TIM1-mediated infection by Ebola, dengue, and West Nile viruses Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission Structure and dynamics of cinnamycinlipid complexes: Mechanisms of selectivity for phosphatidylethanolamine lipids Lanthiopeptin, a new peptide antibiotic. production, isolation and properties of lanthiopeptin Automated comparative protein structure modeling with SWISS-MODEL and Swiss-PdbViewer: A historical perspective SWISS-MODEL: Homology modelling of protein structures and complexes Structural and antimicrobial features of peptides related to myticin C, a special defense molecule from the Mediterranean mussel Mytilus galloprovincialis The SWISS-MODEL Repository-new features and functionality Solution structure of a hydrophobic analogue of the winter flounder antifreeze protein Anti-HSV-1 activity of aspergillipeptide D, a cyclic pentapeptide isolated from fungus Aspergillus sp. SCSIO 41501 Computational peptidology approach to the study of the chemical reactivity and bioactivity properties of aspergillipeptide D, a cyclopentapeptide of marine origin Marine sponges as a drug treasure Bioactive compounds from marine sponges: Fundamentals and applications Mytilus galloprovincialis myticin C: A chemotactic molecule with antiviral activity and immunoregulatory properties Skin antifreeze protein genes of the winter flounder, Pleuronectes americanus, encode distinct and active polypeptides without the secretory signal and prosequences Structural and functional characterization of a multifunctional alanine-rich peptide analogue from Pleuronectes americanus Peptide-lipid interactions: Experiments and applications Small molecules targeting severe acute respiratory syndrome human coronavirus −)-Epigallocatechin-3-gallate is a new inhibitor of hepatitis C virus entry Potential antivirals and antiviral strategies against SARS coronavirus infections Antimicrobial peptides and other peptide-like therapeutics as promising candidates to combat SARS-CoV-2 Antiviral peptides against the main protease of SARS-CoV-2: A molecular docking and dynamics study Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay Challenges for drug repurposing in the COVID-19 pandemic era Drug repurposing in the COVID-19 era: Insights from case studies showing pharmaceutical peculiarities Possibility of HIV-1 protease inhibitors-clinical trial drugs as repurposed drugs for SARS-CoV-2 main protease: A molecular docking, molecular dynamics and binding free energy simulation study Repurposing of drugs for SARS-CoV-2 using inverse docking fingerprints Current strategies of antiviral drug discovery for COVID-19. Front. Mol. Biosci. 2021, 8, 671263 Drug repurposing approach to fight COVID-19 Repurposing drugs for the management of patients with confirmed coronavirus disease 2019 (COVID-19) New hopes for drugs against COVID-19 come from the sea Plitidepsin: Mechanisms and clinical profile of a promising antiviral agent against Covid-19 Plitidepsin: A repurposed drug for the treatment of COVID-19 Plitidepsin: Design, development and potential place in therapy. Drug Des Field-template, QSAR, ensemble molecular docking, and 3D-RISM solvation studies expose potential of FDA-approved marine drugs as SARS-CoVID-2 main protease inhibitors Hunting the main protease of SARS-CoV-2 by plitidepsin: Molecular docking and temperature-dependent molecular dynamics simulations Plitidepsin has potent preclinical efficacy against SARS-CoV-2 by targeting the host protein eEF1A Preclinical and randomized phase I studies of plitidepsin in adults hospitalized with COVID-19 Plitidepsin as a successful rescue treatment for prolonged viral SARS-CoV-2 replication in a patient with previous anti-CD20 monoclonal antibody-mediated B cell depletion and chronic lymphocytic leukemia Exploring privileged structures: The combinatorial synthesis of cyclic peptides Cyclic peptide therapeutics: Past, present and future Natural cyclic peptides as an attractive modality for therapeutics: A mini review Cell-penetrating peptides: 20 years later, where do we stand? Synthetic therapeutic peptides: Science and market Virtual screening of marine natural compounds by means of chemoinformatics and CDFT-based computational peptidology The author thanks the Deutsche Gesellschaft für Internationale Zusammenarbeit (GIZ) for support with the Returning Experts-Migration and Diaspora Program. The author declares no conflict of interest.