Next Article in Journal
Anti-Obesity Effects of a Mixture of Atractylodes macrocephala and Amomum villosum Extracts on 3T3-L1 Adipocytes and High-Fat Diet-Induced Obesity in Mice
Previous Article in Journal
High Resolution Infrared Spectroscopy in Support of Ozone Atmospheric Monitoring and Validation of the Potential Energy Function
Previous Article in Special Issue
Phenylalanine Butyramide Is a New Cosmetic Ingredient with Soothing and Anti-Reddening Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Antiviral Action of Alkaloids

1
Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
2
Department of Environment Technologies and Management, Kuwait University, P.O. Box 5969, Kuwait City 13060, Kuwait
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(3), 903; https://doi.org/10.3390/molecules27030903
Submission received: 1 December 2021 / Revised: 24 January 2022 / Accepted: 26 January 2022 / Published: 28 January 2022
(This article belongs to the Special Issue Lipids and Alkaloids for Therapeutic and Cosmetic Applications)

Abstract

:
Viral infections and outbreaks have become a major concern and are one of the main causes of morbidity and mortality worldwide. The development of successful antiviral therapeutics and vaccines remains a daunting challenge. The discovery of novel antiviral agents is a public health emergency, and extraordinary efforts are underway globally to identify safe and effective treatments for different viral diseases. Alkaloids are natural phytochemicals known for their biological activities, many of which have been intensively studied for their broad-spectrum of antiviral activities against different DNA and RNA viruses. The purpose of this review was to summarize the evidence supporting the efficacy of the antiviral activity of plant alkaloids at half-maximum effective concentration (EC50) or half-maximum inhibitory concentration (IC50) below 10 μM and describe the molecular sites most often targeted by natural alkaloids acting against different virus families. This review highlights that considering the devastating effects of virus pandemics on humans, plants, and animals, the development of high efficiency and low-toxicity antiviral drugs targeting these viruses need to be developed. Furthermore, it summarizes the current research status of alkaloids as the source of antiviral drug development, their structural characteristics, and antiviral targets. Overall, the influence of alkaloids at the molecular level suggests a high degree of specificity which means they could serve as potent and safe antiviral agents waiting for evaluation and exploitation.

Graphical Abstract

1. Introduction

Viral epidemics and pandemic diseases have threatened humanity historically and have had significant impacts on human society [1]. Viruses, followed by some bacteria and protozoa, have been classified as the key pathogens causing human illnesses since 1973 [2]. Viruses are known for serious infectious diseases they cause, including human immunodeficiency virus (HIV), hepatitis B and C (HBV and HCV), coronaviruses including Middle Eastern respiratory syndrome (MERS) and extreme acute respiratory syndrome (SARS), influenza (seasonal), smallpox, viral hemorrhagic fever (Ebola), dengue fever, and chikungunya virus [3,4]. New human virus species are still being detected, at a rate of three or four per year, and over two-thirds of all new human pathogens are viruses. In 2019, there were 690,000 deaths due to AIDS-related illnesses worldwide, compared with 1.7 million in 2014 [5]. A newly detected coronavirus-induced disease (COVID-19) was declared a pandemic in 2020 and has seriously affected the world population, resulting in over 216,074,000 infected cases and 4,496,998 deaths until 27 August 2021, with a notable upward trend [6].
Viruses, particularly the rapidly mutating ones, pose a great pandemic threat to human health especially due to the limited number of vaccines and antiviral drugs. However, many antiviral drugs have been developed since the 1960s and have helped the world to coexist with various viral infections [7]. Over the past 30 years, about 50% of approved medicines have been produced from natural products, either directly or indirectly [8]. This stems from a long tradition of treating various diseases using particular types of plants (herbs) as medical therapeutics.
Alkaloids are a large and structurally diverse group of natural products of microbial, plant, and animal origin. Alkaloids are usually nitrogenous small organic molecules of plant origin, and about 20% of plant species contain alkaloids [9]. Several families of the plant kingdom, such as the Ranunculaceae, Papaveraceae, Fabaceae, Loganiaceae, and Menispermaceae, contain alkaloids [10]. Among other bioactive ingredients of medicinal plants, natural alkaloids have been successfully developed into chemotherapeutic drugs such as camptothecin (CPT), a well-known topoisomerase I inhibitor and vinblastine, which is a highly successful antitumor drug. Similarly, quinidine is used for restoring normal sinus rhythm, treating atrial fibrillation and flutter, as well as ventricular arrhythmias. Another alkaloid named papaverine is used to treat many types of smooth muscle spasms, such as “vascular spasms” associated with acute myocardial infarction and angina pectoris, as well as visceral spasms [11,12]. Alkaloids also exhibit different biological activities, for instance, antitumor, antidepressant, anti-inflammatory, anti-angiogenic, and anti-dementia [13,14,15,16]. In this review, we summarize the experimental evidence for the antiviral activity of natural alkaloids with a half-maximal effective concentration (EC50) or half-maximal inhibitory concentration (IC50) below 10 μM. We also highlight the molecular sites most often targeted by natural alkaloids acting against different virus families.

2. Research Methodology

Pertinent literature was obtained by concomitantly searching the words “bioactive, alkaloid/phytochemicals”, “antiviral”, and “activity” in the Google scholar, Pub Med, Science Direct, Scopus, SpringerLink, Web of Science databases. The literature search was independently performed by one researcher. One hundred forty- two articles from 1950 up to 2021, were selected, cited, and carefully analyzed in order to find data related to the topic and aims of this review. All articles and information used for this review were either published or accepted for publication in English language peer-reviewed journals. Mendeley desktop software was used for preparing the bibliography. The search criteria used were: (1) the antiviral potential of natural alkaloid, (2) isolated phytochemicals alkaloids, or its derivatives effective against DNA and RNA viruses, and (3) antiviral activities of alkaloids that show at a half-maximal effective concentration (EC50) or half-maximal inhibitory concentration below (IC50) below 10 μM with no significant toxicity. However, the data from only 25 investigated pure compounds through in vitro and in vivo studies met the inclusion criteria of this review. Exclusion criteria: (1) antiviral activities with an inhibitory concentration (EC50, IC50) higher than 10μM, (2) literature duplication, (3) non-relevant articles, and (4) studies involving plant extracts with undefined active compounds.

3. Natural Products as a Source for Antiviral Drugs Development

Although vaccinations have been very effective in controlling many viral diseases, some infections can probably be treated only by antiviral chemotherapy. Antiviral drugs could provide the best protection strategy against a virus outbreak which is a permanent threat. Various FDA approved antiviral drugs are available for the inhibition and treatment of virus-related infections, such as rimantadine and amantadine for the inhibition and treatment of influenza A virus, and acyclovir and vidarabine against the varicella-zoster virus and herpes simplex virus infections. Similarly, ribavirin is used against the respiratory syncytial virus and valaciclovir as well as famciclovir against varicella-zoster virus infections. Notwithstanding its benefit, antiviral chemotherapy has some restrictions, namely a narrow antiviral range, ineffectiveness against a dormant virus, and evolution of drug-resistant mutants [7,17].
Natural products have played a significant role in drug development. There could be no accurate and comprehensive description of the role of natural products in drug discovery without the mention of alkaloids. The use of natural products began with the discovery and isolation of morphine from the opium poppy in 1803 [18]. There are several historical examples in which the therapeutic agent present in a natural remedy helped to reveal new physiological features; for instance, morphine enabled an understanding of the opiate receptors and the pathways of endorphin and enkephalin [19]. Recent trends in the pharmaceutical industry have become increasingly focused on small molecules from plants as a source of potential antiviral agents in the context of an urgent need for new therapeutic agents due to the issues of drug resistance and side effects of some drugs [20]. The application of advanced technologies and methods in molecular biology as well as reverse genetics has dramatically increased the possibility of conducting screening of new phytochemical compounds from plant extracts [21].
Since 1952 numerous research efforts have been made to determine the antiviral efficacy of medicinal plants [22]. Subsequent studies have investigated the extracts of several medicinal plants for their antiviral activities. For instance, an investigation into traditional medicinal plants in Togo revealed ten different plants that exhibited antiviral activities against herpes simplex, sindbis virus, and poliovirus [23]. The antiviral activities of nine Malagasy plants have been reported as having a significant impact on the herpes simplex virus (HSV), and four plants have been described as acting against the sindbis virus [24]. A total of 105 plant species have been identified and reported for their potential antiviral activities against a wide range of viruses [25]. The most recently adopted strategy for employing natural products in drug discovery involves moving from a broader approach to a narrower and more applicable one using standard procedures that involve the isolation, characterization, and identification of potent phytochemicals followed by in vitro and in vivo assays to establish the most promising phytochemicals before a clinical trial begins [26]. The commonly isolated and identified phytochemicals from medicinal plants include polyphenols, alkaloids, flavonoids, saponins, quinones, terpenes, proanthocyanins, tannins, polysaccharides, steroids, and coumarins. Among the plant phytochemicals, alkaloids represent the largest antiviral category with a broad spectrum of antiviral properties [3,27,28]. Based on our preliminary independent survey, the alkaloids comprised the largest percentage (22%) of investigated antiviral phytochemicals that were investigated against 15 families of viruses (Data not shown).

4. Structure and Characteristics of Alkaloids

Alkaloids are naturally occurring compounds containing carbon, hydrogen, nitrogen, and oxygen and are present in plant tissues as organic acid salts (e.g., acetic, malic, lactic, citric, or oxalic) and/or weak bases such as nicotine. In addition, some alkaloids may exist as glycosides, such as the solanum (solanine), amides (piperine), and esters (atropine, cocaine) [29]. Alkaloids can also be categorized according to their biosynthetic pathway [30] or their occurrence as heterocyclic or nonheterocyclic alkaloids [31]. They are often classified according to their molecular skeleton, for example., quinoline, indole, isoquinoline, tropane, steroidal, and pyridine and pyrrolizidine alkaloids [32].
Plant-based alkaloids are present in the form of crystalline, amorphous, non-odorous, and non-volatile compounds [31]. They are a very diverse class of secondary metabolites, encompassing an extremely divergent chemical structure. However, the structures of each of the compounds have different functional groups that give the compound its unique characteristics. The skeletal, structural, and functional diversity makes them a versatile group of secondary metabolites, meaning that they are a more significant pharmacophoric unit with desired biological activities [29,33]. According to Lipinski’s criteria, for a molecule to be drug-like, the physicochemical properties of alkaloids (molecular weight, number of NH and OH groups, the average number of N and O atoms per alkaloid, and log P) must fall within the range of the standard criteria for drug-like molecules [33,34]. Alkaloids have a low-to-moderate molecular weight (250–600 Daltons); they are water-soluble and lipid-soluble under acidic/neutral and basic conditions, respectively. Their solubility gives them a unique characteristic that is appropriate for a wide range of medical uses, as they can be transported in the protonated form and penetrate cell membranes in the neutral form. The pKa values of alkaloids range from 6 to 12 [32,35].

5. Targets for Antiviral Alkaloids

More recently, significant progress has been made in the screening and use of small molecules (with low molecular weight) as inhibitors of biochemical processes. Natural products, especially alkaloids, are small molecules potentially useful for this method of investigation [36], as they have been proven to interact with a wide range of biological targets. Their mechanism of action varies with their class and chemical structure [37]. Both isolated compounds and plants (or extracts thereof) containing alkaloids have been widely investigated and used. Alkaloids have been proven to interact with a wide range of biological targets [37]. Antioxidant properties, scavenging capabilities, DNA and RNA synthesis inhibition, and viral replication blockage are the main antiviral activities of many adopted phytochemical compounds, and these activities may be due to the synergistic effects of more than one mechanism [38]. It has been extensively stated that there is a significant association between the concentration and toxicity of any substance used as a drug, and lower doses of drugs are always recommended. In our preliminary literature review, we found that alkaloids have dose-dependent effects, and most of the studies carried out at concentrations lower than 10 µM showed effective antiviral activities with no significant toxicity. Thus, reviewing the acceptable practices for studying antiviral activities of alkaloids, we focused on the antiviral effects of alkaloids at concentrations in the order of magnitude of 10−3–10 µM, which are expected to be pharmacologically more suitable. Furthermore, we subdivided the reviewed alkaloids into two classes (viral attachment and cell entry target and viral replication target), based on the mechanisms of action. Experimental evidence of the interaction and interference of alkaloids with cellular components is summarized in Table 1.

5.1. Viral Attachment and Cell Entry Target

Viral cell entry is the first essential step in the viral life cycle and infection establishment since viral infection requires the transfer of the viral genome into the host cell cytoplasm, either at the plasma membrane or the endomembrane system. Virus entry and post-entry stages have been the subject of several advanced recent studies. The prevention of viral infections through the host cell is widely considered to be one of the appealing targets of effective drug development. Moreover, host cell targeting could reduce drug resistance as the target compounds would act on the host proteins [72,73]. For both enveloped and non-enveloped viruses, entry into the cell begins with an attachment to cell-surface receptors and end with the delivery of the viral genome to the cell cytoplasm or nucleus, where its replication process can start. This can occur either by fusion (for enveloped viruses) or penetration (for non-enveloped viruses) [74,75].
The entry of enveloped viruses typically involves fusion between the viral lipid envelope and the host membrane. Growing evidence suggests that alterations in the lipid membrane can block viral release and entry. The alterations can be caused by fusion inhibitors, a class of antiviral drugs that act extracellularly to prevent direct attachment and entry of viruses. They include certain lipid active compounds, a potential antiviral agent [76]. For example, LJ001 is a membrane-binding molecule that was proven to effectively inhibit the viral entry of numerous enveloped viruses. Through its nonpolar end, LJ001 penetrates viral lipid membranes and inhibits viral entry, possibly by affecting the rigidity and curvature of the membranes, thus preventing efficient virus–cell membrane fusion [77].
Virus entry is specific for host cell susceptibility and depends on viral surface proteins and receptors. The enveloped viruses use a two-step procedure to release their genetic material into the cell, typically they first bind to specific surface receptors of the target cell membrane and then fuse between the viral lipid envelope and the host membrane. Recent studies revealed that alterations in the lipid membrane can block viral release and entry, suggesting their potential as antiviral drugs. Like some other small-molecule inhibitors, the alkaloid michellamine B has previously been shown to suppress HIV infection at an early stage by inhibiting cellular fusion and syncytium formation [63]. Further, several related compounds have been identified as acting by blocking the attachment and entry of some novel viruses. For example, emetine was identified as an entry inhibitor of the Zika virus (ZIKAV) and Ebola (EBOV) viruses, disrupting lysosomal function and blocking the ZIKAV NS5 polymerase activity [48]. In ZIKAV, the fusion loop epitope, a segment of the envelope (E) glycoprotein, represents a potential therapeutic target impacting host attachment [78]. In filoviruses such as EBOV, the spike glycoprotein (S) promotes membrane fusion and plays an essential role in virus entry and contact with the cell [79].
HIV entry also involves fusion between the viral lipid envelope and the host cell. Thus, fusion inhibitors can prevent direct attachment and entry of HIV. Mcmahon et al. reported the inhibition of the HIV life cycle by michellamine B at an early stage by inhibiting RT and at later stages by inhibiting cellular fusion and syncytium formation. The same authors also revealed that michellamine B inhibited the production of RT, p24, and infectious virions in HIV-infected CEM-SS cells and freshly isolated peripheral blood cells. The inhibition of cell fusion by michellamine B may result from reversible interactions with membrane components that play a role in cell–cell fusion and focus on the entry process due to the amphoteric nature of alkaloids [63]. Besides, in another study, infection by the RT M184V HIV-1 mutant (in reverse transcriptase, RT) was successfully blocked by emetine, thereby showing a direct inhibitory effect on viral entry. It appears that the underlying mechanism of this inhibition may involve interference with intact HIV particles and block the reverse transcription reaction, which often prevents the entry of a virus to the host [48]. Emetine was also identified as an entry inhibitor of the Zika (ZIKAV) and Ebola virus diseases (EVD) by disrupting lysosomal function and blocking the ZIKAV NS5 polymerase activity [48]. Further, several related compounds have been identified as effective by blocking the attachment and entry of some novel viruses. In a recent study, a full agreement of molecular docking simulation with the experimental results was observed for the binding of berberine to the E2 glycoprotein of HCV and specific blocking of the viral attachment and entry [80]. In the case of SARS-CoV-2, spike (S) protein S2 subunit plays a key role in mediating virus fusion with and entry into the host cell. The heptad repeat 1 (HR1) and heptad repeat 2 (HR2) of the S protein can interact to form a six-helical bundle (6-HB), thereby bringing viral and cellular membranes in close proximity for fusion [81]. Recent computational studies showed that piperine and berberine alkaloids have a significant binding affinity towards the spike glycoprotein of SARS-CoV-2 and ACE2 receptor and may be useful as a therapeutic agent for restricting viral attachment to the host cells [82]. Another molecular docking and dynamic simulation study predicted a potential inhibitory effect of alkaloids on SARS-CoV-2 infection by binding to the receptor-binding domain of the SARS-CoV-2 S protein and putatively preventing it from binding to the host cell. Therefore, based on the simulation investigations, it was suggested that the alkaloids (cryptospirolepine, 10-hydroxyusambarensine, and cryptoquindoline) might have the potential to prevent SARS-CoV-2 infection [83,84]. However, their applicability as antiviral compounds against SARS-CoV-2 needs to be studied experimentally to provide strong evidence of the interaction and inhibition.
Several studies suggested that many natural alkaloids effectively interfere with the cell membrane (even though this membrane is not the final target) and indirectly function by binding to the viral glycoprotein. The amphiphilic nature of alkaloids is likely to provide a rationale for their antiviral activities, particularly as an entry inhibitor preventing viral attachment. Chemically, amphiphilic alkaloids have a lipophilic molecular framework and a specific functional group (Figure 1). Amphiphilic alkaloids have a predominantly hydrophobic character that enables permeation of the cell membrane and then interaction with the membrane components such as proteins or lipids at the periphery of the cells. They can also interact with various receptors and ion channels, resulting in physicochemical property changes of the lipid bilayer and in the activities of different receptors, ion channels, and enzymes [85,86,87,88,89]. Research on alkaloid–cell membrane interaction explained the possible mechanism of alkaloids penetration through the cell membrane. The results suggested that the alkaloid–lipid interaction affects the lipid headgroup structure by weakening the lipid headgroup–headgroup H bonds and enhancing the electrostatic attraction between the alkaloids and the lipid phosphate groups, facilitating the alkaloid passage between the headgroups [85].
However, the cell membrane is the main target site of specific fusion alkaloid inhibitors leading to the disruption of the glycoprotein within the virion envelope, surface receptors, and the disruption of the attachment of viruses to host cells, reducing the possibility of effective virus–host interaction [90,91,92,93,94]. Therefore, conformational rearrangements of the viral membrane lipids strongly influence viral membrane fusion and may even be exploited for the design of broad-spectrum antivirals against a wide variety of enveloped viruses.

5.2. Viral Replication Target

As the nucleic acids are the most desirable target for inhibiting the specific viral replication step, therefore, research on their structural and conformational changes resulting from their interactions with small molecules is currently a matter of intense interest [95]. Many alkaloids attack the genetic material and enzymes involved in the viral life cycle through multiple mechanisms and interrupt their natural biological functions. In this section, we review the antiviral activities of natural alkaloids at the molecular level, specifically targeting the synthesis of DNA and RNA, as well as proteins.

5.2.1. Inhibition of DNA and RNA Synthesis

Viruses vary in form and complexity. They consist of genetic material, DNA or RNA, either of which may be single or double-stranded, surrounded by a coat composed of protein. Viruses encode several functions necessary for their life cycle. However, they are entirely dependent on the protein synthesis machinery of their host cells [96]. First, viruses use cellular ribosomes to translate their viral mRNA into structural proteins, envelope proteins, and viral enzymes [97]. RNA viruses replicate in the cytoplasm and rely on their RNA genome as a template for the synthesis of additional RNA. Three types of RNA must be synthesized during the replication process, including the genome, a copy of the genome, and mRNAs [98]. Viral mRNA transcripts of DNA viruses are produced within the cell nucleus through the host’s DNA-dependent RNA polymerase II. Replication of a DNA virus is highly accurate as the DNA polymerase checks the copied sequences (proofreading) and removes most mismatches [97].
The replication cycle of viruses is complex and generally involves three main steps: initiation of the infection, genome replication and expression, and finally, the release of mature virions from the infected cell [99]. Replication cycles during the viral life cycle represent apparent targets for intervention. Cumulative evidence suggests that HIV reverse transcriptase is the major anti-HIV target involved in viral replication [100]. Chain termination inhibiting the herpes virus replication process [101] is another example, as well as polyamine depletion, which restricts replication, translation, and packaging of diverse RNA and DNA viruses [102,103]. Viral proteases are very successful, and their inhibitors are actively developed [104]. Current evidence suggests that RNA-dependent RNA polymerase could be a target for the inhibition of SARS CoV-2 [105].
Inhibiting nucleic acid synthesis sounds like a great strategy for antiviral activity. Nucleic acid intercalation is a well-known process for interrupting DNA-associated pathways as they perturb DNA structure, which can, in turn, influence DNA processing. This alteration in structural and mechanical properties of DNA could offer broad-spectrum antiviral agents that are more effective than target-specific compounds in preventing multiple emerging infectious diseases. Generally, DNA–ligand H noncovalent interactions require the presence and insertion of an appropriate size and chemical nature of moieties between adjacent DNA base pairs [106]. These ligands are mostly aromatic, planar, and polycyclic and, therefore, often make good nucleic acid stains/dyes [107]. DNA and RNA intercalation is of great interest in the context of many alkaloids and represents a good inhibition activity. A series of studies combining biochemical methods with numerous interdisciplinary techniques provided clear evidence on alkaloid–nucleic acid interaction effects. It has been previously established in trypanosomes that some of the alkaloids can effectively intercalate with DNA or DNA processing enzymes such as DNA polymerase I and reverse transcriptase. Other alkaloids have profound effects on the RNA polymerases and DNA topoisomerase, thus impairing base-pairing and blocking DNA replication, DNA repair, and transcription, which remain a vital drug target [108]. Many of these intercalators have planar geometry with aromatic moieties that can smoothly fit between AT and GC base pairs [109]. Intensively studied DNA-alkaloids intercalators include berberine, emetine, sanguinarine, β-carboline, quinine, skimmianine, cinchonine, and dictamine [29]. The intercalation of berberine and palmatine alkaloid molecules within a triple-helical RNA structure, poly(U), poly(A), poly(U), was experimentally demonstrated using various biophysical and calorimetric techniques. This study brought an advanced understanding of the specific binding interaction of alkaloids to RNA structures, particularly triplexes, and may be useful in developing RNA-targeted therapeutics [110]. Although DNA–alkaloid intercalation has been extensively studied and successfully demonstrated as a broad biological activity in different organisms, only a few studies have been carried out on the antiviral effects. The reviewed studies suggested that intercalation of an alkaloid, such as buchapine, colchicine, acronycine, and lycorine, into DNA, could serve as a promising inhibitory mechanism against HIV, HSV, and human cytomegalovirus (HCMV) replication, via structural changes in the DNA that could lead to irreparable damage in the genetic material of the virus [47,111,112].
The capacity of several alkaloids to inhibit DNA and RNA synthesis in multiple virus families via different mechanisms has been experimentally proven (Table 1). For example, oliverine interferes with and inhibits HSV-1 at the early stage of replication via the suppression of DNA synthesis [63]. The lycorine inhibits the viral RNA-dependent polymerase (RdRp) of ZIKAV and dengue virus (DENV) [60]. Emetine exhibits remarkable inhibitory activity by blocking the HIV reverse transcriptase and reaching an 80% reduction in HIV-1 infection, with a low cytotoxic effect [49]. Emetine also inhibited viral polymerases [86] and the RNA synthesis of DNA and RNA viruses such as DENV, buffalopoxvirus (BPXV), bovine herpesvirus 1 (BHV-1), and Newcastle disease virus (NDV) without generating drug-resistant virus variants. This suggests that emetine could provide significant therapeutic value against some of these viruses without inducing an antiviral drug-resistant phenotype [47,113]. Tomatidine is a steroidal alkaloid derived from the stem and leaves of unripe, green tomatoes. It has been recently described as having antiviral activity towards CHIKV infection. This is commonly achieved by interfering with the production of infectious viral particles and inhibiting replication after virus attachment and cell entry [69]. Camptothecin is a DNA topoisomerase 1 (TOP1) inhibitor that inhibits enterovirus 71 (EV71) RNA replication and translation, altering the template DNA and thus affecting the synthesis of DNA and RNA. Alternatively, camptothecin may inhibit the activity of an enzyme involved in DNA repair, such as DNA ligase degrading the DNA, thereby representing irreparable DNA damage [44].

5.2.2. Inhibitors of Protein Synthesis

Targeting the virus life cycle at the protein synthesis level with alkaloids demonstrates the molecular basis of specificity in nucleic acid–alkaloid interactions, for example, the associations of alkaloids with different nucleic acids, such as ribosomal ribonucleic acid (rRNA), transfer ribonucleic acid RNA(tRNA), or messenger RNA (mRNA) are different and specific. Some alkaloids were found to be specifically toxic to plasmodia, as they prevented the biosynthesis of plasmodial DNA by preventing the integration of building blocks into plasmodial DNA [114,115]. A typical example of this group of alkaloids is omacetaxine. Omacetaxine is a natural homoharringtonine alkaloid from the Cephalotaxus genus that has been approved by the FDA as a protein synthesis inhibitor drug [116].
Several alkaloids have been proven to be potent inhibitors of protein synthesis in various families of viruses. A likely mode of action is blocking the ribosomal protein. The 40S ribosomal subunit appears to be the site of a specific receptor for emetine, which inhibits viral translation [117,118]. Other studies have reported the multiple actions of alkaloids, for example, emetine, which can block the reverse transcriptase of HIV, and inhibit viral polymerases, in addition to suppressing the viral translation by disrupting the 40S ribosomal protein S14 and blocking the synthesis of the viral proteins in infected cells [47,51,109,113]. Fresno et al. reported that some alkaloids can block the formation of peptide bonds and inhibit the enzyme binding of aminoacyl-tRNA [119].
As summarized in Table 1, the effect of alkaloids on specific steps of the viral life cycle has been widely reported. For example, a possible inhibitory mechanism of emetine on viral genome replication could be due to direct inhibition of viral protein synthesis. Since virus replication requires the presence of viral structural and non-structural proteins, their inhibition may eventually block viral genome replication [49]. Efficacy and mechanism of low dose emetine as a human cytomegalovirus inhibitor in a mouse model (HCMV) was also reported. The inhibition of HCMV by emetine occurred after virus entry but before DNA replication and resulted in a decreased expression of viral proteins, which are essential for the production of infectious HCMV. Synergistic virus inhibition was achieved when emetine was combined with ganciclovir [51]. Emetine has also been shown to block the synthesis of the viral proteins in HIV-1, suggesting the importance of further studies to evaluate the potential use of this compound as an anti-HIV inhibitor. Earlier studies demonstrated the inhibition of encephalomyocarditis virus protein synthesis by emetine and harringtonine alkaloids. The observed inhibition of protein synthesis occurred during the elongation step [120]. Similarly, polypeptide chain elongation was blocked by the interaction of lycorine with the C-terminal region in the polyproteins during the elongation step, and lycorine thus inhibited protein synthesis of EV71 [121]. Lycorine inhibits protein synthesis in eukaryotic cells by inhibiting the peptide bond formation step [122]. Lycorine inhibits the transpeptidation reaction by suppressing the coupling of the N-acetyl leucyl residue from UACCA-acetyl leucyl to puromycin [123]. Berberine inhibits protein synthesis through viral protein trafficking or maturation and TNF-α and PGE2 [42]. Berberine also interferes with the viral immediate-early 2 (IE2) protein of cytomegalovirus and remarkably inhibits the synthesis of the proteins essential for regulating the expression of many host genes and the production of infectious HCMV [40]. Quinine significantly suppresses HSV-1 protein synthesis, inhibits early steps of the influenza A virus (H1N1) duplication cycle [124], inhibits DENV by decreasing viral RNA and protein synthesis, and amends the expression of the genes connected to inborn immune response [125]. Alkaloid antiviral effects dramatically prevent the completion of the viral growth of different strains in infected cells. Most of these phytochemicals attack cellular enzymes and viral proteins. The findings suggest that alkaloids target the cellular and viral components with multiple action mechanisms, acting as specific or broad-spectrum inhibitors of viral replication. Every essential step in the virus life cycle is an attractive target for antiviral alkaloids. Remarkably, the antiviral activities of alkaloids inhibit replication by targeting cellular enzymes and protein synthesis. Some alkaloids have dual cellular mechanisms, such as fusion and replication inhibition. In addition, some alkaloids show specific inhibition of protein synthesis, mostly at the peptide bond formation step. Most studies confirmed protein synthesis inhibition through action on the RdRp 40S ribosomal subunit and some other specific virus receptors. Ultimately, the interference of alkaloids with nucleic acids resulted in an alteration in the nucleic acid sequence, leading to blockage of the transfer of messages important for protein synthesis, thereby preventing the protein production required for many viral infections.

6. Toxicology and Side Effects

Irrespective of their established antiviral activities, some alkaloids are recognized as too toxic and are banned for animal and human use. The harmfulness of alkaloids, as with other drugs, depends on the dose. However, it is difficult to make conclusions about the overall significance and utilization of alkaloid compounds considering the lack of specific data on the toxicokinetics of the examined alkaloid compounds in clinical studies. Nevertheless, the compounds highlighted in this review displayed no harmful effects in in-vitro studies at lower concentrations. Thus, in the next section, attention will be paid to the major restrictions of the studied alkaloids in terms of their toxicity in preclinical and clinical trials.
As described above, camptothecin (CPT) has a confirmed and efficient antiviral activity. The main disadvantage of using CPT as an antiviral agent is its cytotoxicity, found in HT-29 cells with an IC50 value of 10 nM. CPT induces DNA impairment at concentrations as low as 51 nM in whole cells and 12 nM in isolated nuclei in in-vitro studies. Furthermore, results from clinical trials in the treatment of progressive gastrointestinal cancer were not as promising and were discontinued in the 1970s [126]. Attention will certainly remain focused on camptothecin due to both the present pharmaceutical possibilities and the additional analogs or derivatives of camptothecin. New progress in CPT-related antiviral molecules represents the synthesis of camptothecin associated analogs with notably low cytotoxicity [55,127]. A new camptothecin-20-O-propionate hydrate (CZ48) was created and presented as an anticancer agent against 19 human tumor xenografts, lymphoma, and solid tumors [128]. No toxicity was experienced in the in-vitro study. As a result of the harmless nature of the drug in rats, the highest tolerated dose was not reached [119]. Continuing clinical safety-related trials were carried out in a single-arm, non-randomized viability and phase I trial of 20 camptothecin propionate administered run orally to 65 participants at the Cancer Therapy and Research Center at the UT Health Science Center at San Antonio, Texas [129]. A clearer picture of these side effects and the toxicity will be known once the results of these clinical trials are released. A recent in vitro investigation of nanoencapsulated camptothecin prevented HCMV replication at a low concentration suggesting that the delivery method could alter the toxicity [130]. The extensiveness of preclinical and clinical data on CPT analogs will be needed to help direct the preparation and scientific development of a lead therapeutic compound.
Michellamine B is the major in vitro active anti-HIV component. An earlier update on plant-derived components used for the treatment of HIV based on preclinical assays showed that the effective antiviral activities of michellamine B could only be attained at close to neurotoxic dose levels. For example, an effective dose of 25 mg per kg has been regarded as too toxic for clinical trials, and therefore the additional studies for the treatment of HIV infections in the United States were suspended [131]. Notwithstanding the cessation of the development of michellamine B as a possible anti-HIV agent, there is still the potential to develop less toxic and simpler analogs of michellamine B, which are currently being widely examined [132,133].
One of the observed examples of alkaloid intercalating into DNA is colchicine which was approved by the FDA in 1961 and is generally used in the management of gout [134]. Colchicine may exert an immuno-suppressive effect. However, the tolerability and safety of colchicine have been confirmed in huge randomized cardiovascular trials [135]. There are now 28 continuing clinical trials of colchicine to examine its positive effects on moderate to severe COVID-19 infections [136]. A phase 3, randomized, double-blind, placebo-controlled multicenter study to assess the safety and efficacy of colchicine in 4065 adult patients diagnosed with COVID-19 infection is continuing, and as the outcome of these trials become accessible, it should become clearer whether there is a benefit to using colchicine in patients with COVID-19. Another recently completed study comprising 36 COVID-19 patients showed that colchicine reduced the length of both additional oxygen therapy and hospitalization, and the drug was safe and well-tolerated [137].
Emetine is an alkaloid of substantial medicinal value. Despite its antiviral strength, its curative use has several side effects, and the use of the drug has been diminished by dose-dependent toxicity. Cardiotoxicity is the most dangerous and serious hostile effect of emetine. The clinical indications are hypotension, dysrhythmias, tachycardia, and cardiomyopathy. Electrocardiographic anomalies happen in 60–70% of cases [138,139]. A study looking at emetine dihydrochloride hydrate as a possible candidate for use against malaria was recently undertaken. In this study, a possible route to connect the nanomolar (median effective dose, ED50, of 47 nM) antimalarial efficiency of this inexpensive natural product was described to minimize the formerly reported dose-related toxicity of the drug [140].
Lycorine is the most common Amaryllidaceae alkaloid and has an extensive array of bioactivities. In disparity with its possible health-promoting effects, it is assumed to be the cause of plant poisoning in animals and humans and exhibits numerous side effects. Results proved that lycorine at 1.0 mg/kg body weight causes emesis and nausea in animals and humans due to poisoning [141]. Lycorine as a potential natural candidate for an anticancer drug, was efficient in a very low, single-digit micromolar concentration; the IC50 value generally did not exceed 7.5 μM and it was highly tolerable with negligible toxicity [142,143].
Omacetaxine, a protein synthesis inhibitor, is used for treating leukemia. In a second phase of clinical trials, omacetaxine was tested on 103 patients with chronic myeloid leukemia (CML) with the BCR-ABL T315I mutation, and results showed that omacetaxine might offer an effective and safe treatment for these patients. Omacetaxine has been found to improve persistence in patients with persistent CML and resistance to many tyrosine kinase inhibitors and was therefore accepted for usage in the United States in 2012 [144,145].

7. Conclusions

Much research has been dedicated to combating the pandemic threat of viruses, which can have devastating effects on humans, plants, and animals. Viruses have initiated the deadliest and most horrifying diseases the world has seen. The most recent example of this is the ongoing coronavirus disease 2019 (COVID-19) pandemic. With the rapid development of new resistant viral strains, there is a need to identify antivirals that target these strains with more effectiveness and less toxicity.
Natural plant research has previously resulted in the development of many effective drugs. Some alkaloids are already used in well-known medicines with known action mechanisms and expected health effects. There are many approved alkaloid-derived drugs for different diseases (including infectious ones), such as the antimalarial drug hydroxychloroquine. However, the principal reason that some alkaloids are presumed to be promising antiviral candidates could be their molecular basis of specificity and their ability to target multiple virus families with different mechanisms of action. Therefore, a great deal of work has been carried out to explain the mechanisms of action and the effective and lethal doses of alkaloids to investigate and expand their use as antiviral drugs. These brief, reliable, and replicable scientific facts, along with the current availability of state-of-the-art analytical technology, should be explored translated into actionable clinical trials of natural products.

Author Contributions

Conceptualization, F.L.A., B.S.A.-A. and A.U.; methodology, F.L.A. and A.U.; writing—original draft preparation, F.L.A. and A.U. reviewed the original draft and addressed the reviewers’ comments. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest

Abbreviations

CHIKVChikungunya virus
CVB3Coxsackievirus B3
DENVDengue virus
EV71Enterovirus 71
EVDEbola virus disease
H1N1Influenza A virus
H5N1Asian avian influenza A virus
H7N7Avian influenza A virus
HCMVHuman cytomegalovirus
HCVHepatitis C virus
HIVHuman immunodeficiency virus
HSVHerpes simplex virus
MERSMiddle East respiratory syndrome
MLVMurine leukemia viruses
MMTVMouse mammary tumor viruses
SARSSevere acute respiratory syndrome
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
VACVVaccinia virus
ZIKAVZika virus

References

  1. WHO. Global HIV & AIDS Statistics—2020 Fact Sheet. 2019. Available online: https://www.unaids.org/en/resources/fact-sheet#:~:text=In 2019%2C around 690 000,1.6 million%5D people in 2010 (accessed on 25 April 2020).
  2. Desselberger, U. Emerging and re-emerging infectious diseases. J. Infect. 2000, 40, 3–15. [Google Scholar] [CrossRef] [PubMed]
  3. Krishnan, N.; Devadasan, V.; Raman, P. Plant-derived alkaloids as anti-viral agents. Int. J. Res. Pharm. Sci. 2020, 11, 6174–6182. [Google Scholar] [CrossRef]
  4. Martinez, J.P.; Sasse, F.; Brönstrup, M.; Diez, J.; Meyerhans, A. Antiviral drug discovery: Broad-spectrum drugs from nature. Nat. Prod. Rep. 2015, 32, 29–48. [Google Scholar] [CrossRef] [PubMed]
  5. Woolhouse, M.; Gaunt, E. Ecological Origins of Novel Human Pathogens. Crit. Rev. Microbiol. 2007, 33, 231–242. [Google Scholar] [CrossRef]
  6. Worldometers. COVID-19 Coronavirus Outbreak. Worldometers. 2020. Available online: https://www.worldometers.info/coronavirus/#countries (accessed on 27 August 2021).
  7. De Clercq, E. Chemotherapy of Viral Infections. In Medical Microbiology, 4th ed.; Baron, S., Ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
  8. Newman, D.J.; Cragg, G.M. Natural Products As Sources of New Drugs over the 30 Years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [Green Version]
  9. Ain, Q.-U.; Khan, H.; Mubarak, M.S.; Pervaiz, A. Plant Alkaloids as Antiplatelet Agent: Drugs of the Future in the Light of Recent Developments. Front. Pharmacol. 2016, 7, 292. [Google Scholar] [CrossRef] [Green Version]
  10. Lu, J.-J.; Bao, J.-L.; Chen, X.-P.; Huang, M.; Wang, Y.-T. Alkaloids Isolated from Natural Herbs as the Anticancer Agents. Evid.-Based Complement. Altern. Med. 2012, 2012, 921430. [Google Scholar] [CrossRef] [Green Version]
  11. Huang, M.; Gao, H.; Chen, Y.; Zhu, H.; Cai, Y.; Zhang, X.; Miao, Z.; Jiang, H.; Zhang, J.; Shen, H.; et al. Chimmitecan, a Novel 9-Substituted Camptothecin, with Improved Anticancer Pharmacologic Profiles In vitro and In vivo. Clin. Cancer Res. 2007, 13, 1298–1307. [Google Scholar] [CrossRef] [Green Version]
  12. Li, W.; Shao, Y.; Hu, L.; Zhang, X.; Chen, Y.; Tong, L.; Li, C.; Shen, X.; Ding, J. BM6, a new semi-synthetic vinca alkaloid, exhibits its potent in vivo anti-tumor activities via its high binding affinity for tubulin and improved pharmacokinetic profiles. Cancer Biol. Ther. 2007, 6, 787–794. [Google Scholar] [CrossRef] [Green Version]
  13. Alasvand, M.; Assadollahi, V.; Ambra, R.; Hedayatinezhad, E.; Kooti, W.; Peluso, I. Antiangiogenic Effect of Alkaloids. Oxidative Med. Cell. Longev. 2019, 2019, 9475908. [Google Scholar] [CrossRef]
  14. Gupta, A.P.; Pandotra, P.; Kushwaha, M.; Khan, S.; Sharma, R.; Gupta, S. Alkaloids: A source of anticancer agents from nature. In Studies in Natural Products Chemistry; Elsevier B.V.: Amsterdam, The Netherlands, 2015; Volume 46, pp. 341–445. [Google Scholar]
  15. Peng, J.; Zheng, T.-T.; Li, X.; Liang, Y.; Wang, L.-J.; Huang, Y.-C.; Xiao, H.-T. Plant-Derived Alkaloids: The Promising Disease-Modifying Agents for Inflammatory Bowel Disease. Front. Pharmacol. 2019, 10, 351. [Google Scholar] [CrossRef] [Green Version]
  16. Yan, T.; Xu, M.; Wan, S.; Wang, M.; Wu, B.; Xiao, F.; Bi, K.; Jia, Y. Schisandra chinensis produces the antidepressant-like effects in repeated corticosterone-induced mice via the BDNF/TrkB/CREB signaling pathway. Psychiatr. Res. 2016, 243, 135–142. [Google Scholar] [CrossRef]
  17. Burrell, C.J.; Howard, C.R.; Murphy, F.A. Antiviral Chemotherapy. In Fenner and White’s Medical Virology; Academic Press: Cambridge, MA, USA, 2017; pp. 169–183. [Google Scholar] [CrossRef] [Green Version]
  18. Siddiqui, A.A. Role of Natural Products in Drug Discovery Process. Int. J. Drug Dev. Res. 2014, 6, 172–204. [Google Scholar]
  19. Rishton, G.M. Natural Products as a Robust Source of New Drugs and Drug Leads: Past Successes and Present Day Issues. Am. J. Cardiol. 2008, 101, S43–S49. [Google Scholar] [CrossRef]
  20. Andersen, D.O.; Weber, N.D.; Wood, S.G.; Hughes, B.G.; Murray, B.K.; North, J.A. In vitro virucidal activity of selected anthraquinones and anthraquinone derivatives. Antivir. Res. 1991, 16, 185–196. [Google Scholar] [CrossRef]
  21. Li, J.W.-H.; Vederas, J.C. Drug Discovery and Natural Products: End of an Era or an Endless Frontier? Available online: http://science.sciencemag.org/ (accessed on 21 August 2021).
  22. Chantrill, B.H.; Coulthard, C.E.; Dickinson, L.; Inkley, G.W.; Morris, W.; Pyle, A.H. The Action of Plant Extracts on a Bacteriophage of Pseudornonas pyocyanea and on Influenza A Virus. Microbiology 1952, 6, 74–84. [Google Scholar]
  23. Anani, K.; Hudson, J.B.; De Souza, C.; Akpagana, K.; Tower, G.H.N.; Arnason, J.T.; Gbeassor, M. Investigation of medicinal plants of Togo for antiviral and antimicrobial activities. Pharm. Biol. 2000, 38, 40–45. [Google Scholar] [CrossRef]
  24. Hudson, J.; Lee, M.; Rasoanaivo, P. Antiviral Activities in Plants Endemic to Madagascar. Pharm. Biol. 2000, 38, 36–39. [Google Scholar] [CrossRef]
  25. Sohail, M.N.; Rasul, F.; Karim, A.; Kanwal, U.; Attitalla, I.H. Plant as a Source of Natural Antiviral Agents. Asian J. Anim. Vet. Adv. 2011, 6, 1125–1152. [Google Scholar] [CrossRef] [Green Version]
  26. Koparde, A.A.; Doijad, R.C.; Magdum, C.S. Natural products in drug discovery. In Pharmacognosy—Medicinal Plants; Perveen Al-Taweel, A., Ed.; IntechOpen: London, UK, 2019. [Google Scholar] [CrossRef] [Green Version]
  27. Kapoor, R.S.; Sharma, B.; Kanwar, S.S. Antiviral Phytochemicals: An Overview. Biochem. Physiol. Open Access 2017, 6, 1–7. [Google Scholar] [CrossRef]
  28. Moradi, M.T.; Karimi, A.; Lorigooini, Z. Alkaloids as the natural anti-influenza virus agents: A systematic review. Toxin. Rev. 2018, 37, 11–18. [Google Scholar] [CrossRef]
  29. Wink, M. Inrference of alkaloids with neuroreceptors and ion channels. Stud. Nat. Prod. Chem. 2000, 21, 3–122. [Google Scholar] [CrossRef]
  30. Ramawat, K.G.M.J. Biotechnology: Secondary Metabolites. Science, 2nd ed.; Science Publishers: Enfield, UK, 2007. [Google Scholar]
  31. Kukula-Koch, W.A.; Widelski, J. Chapter 9—Alkaloids. In Pharmacognosy; Badal, S., Delgoda, R., Eds.; Academic Press: Boston, MA, USA, 2017; pp. 163–198. [Google Scholar]
  32. Verpoorte, R. Alkaloids. In Encyclopedia of Analytical Science; Elsevier: Amsterdam, The Netherlands, 2005; pp. 56–61. [Google Scholar]
  33. Cordell, G.A.; Quinn-Beattie, M.L.; Farnsworth, N.R. The potential of alkaloids in drug discovery. Phytother. Res. 2001, 15, 183–205. [Google Scholar] [CrossRef]
  34. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development q Settings. 2001. Available online: www.elsevier.com/locate/drugdeliv (accessed on 15 May 2021).
  35. Ramanathan, S.; Parthasarathy, S.; Murugaiyah, V.; Magosso, E.; Tan, S.C.; Mansor, S.M. Understanding the Physicochemical Properties of Mitragynine, a Principal Alkaloid of Mitragyna speciosa, for Preclinical Evaluation. Molecules 2015, 20, 4915–4927. [Google Scholar] [CrossRef] [Green Version]
  36. Stockwell, B.R. Exploring biology with small organic molecules. Nature 2004, 432, 846–854. [Google Scholar] [CrossRef] [Green Version]
  37. Rao, K.N.; Venkatachalam, S.R. Inhibition of Dihydrofolate Reductase and Cell Growth Activity by the Phenanthroindolizidine Alkaloids Pergularinine and Tylophorinidine: The In Vitro Cytotoxicity of These Plant Alkaloids and their Potential as Antimicrobial and Anticancer Agents. Available online: www.elsevier.com/locate/toxinvit (accessed on 15 May 2021).
  38. Naithani, R.; Mehta, R.G.; Shukla, D.; Chandersekera, S.N.; Moriarty, R.M. Antiviral Activity of Phytochemicals: A Current Perspective. In Dietary Components and Immune Function; Humana Press: Totowa, NJ, USA, 2010; pp. 421–468. [Google Scholar] [CrossRef]
  39. Quintana, V.; Selisko, B.; Brunetti, J.; Eydoux, C.; Guillemot, J.; Canard, B.; Damonte, E.; Julander, J.; Castilla, V. Antiviral activity of the natural alkaloid anisomycin against dengue and Zika viruses. Antivir. Res. 2020, 176, 104749. [Google Scholar] [CrossRef]
  40. Luganini, A.; Mercorelli, B.; Messa, L.; Palù, G.; Gribaudo, G.; Loregian, A. The isoquinoline alkaloid berberine inhibits human cytomegalovirus replication by interfering with the viral Immediate Early-2 (IE2) protein transactivating activity. Antivir. Res. 2019, 164, 52–60. [Google Scholar] [CrossRef]
  41. Song, S.; Qiu, M.; Chu, Y.; Chen, D.; Wang, X.; Su, A.; Wu, Z. Downregulation of Cellular c-Jun N-Terminal Protein Kinase and NF-κB Activation by Berberine May Result in Inhibition of Herpes Simplex Virus Replication. Antimicrob. Agents Chemother. 2014, 58, 5068–5078. [Google Scholar] [CrossRef] [Green Version]
  42. Cecil, C.E.; Davis, J.; Cech, N.B.; Laster, S.M. Inhibition of H1N1 influenza A virus growth and induction of inflammatory mediators by the isoquinoline alkaloid berberine and extracts of goldenseal (Hydrastis canadensis). Int. Immunopharmacol. 2011, 11, 1706–1714. [Google Scholar] [CrossRef]
  43. McCormick, J.L.; McKee, T.C.; Cardellina, J.H.; Boyd, M.R. HIV Inhibitory Natural Products. 26. 1 Quinoline Alkaloids from Euodia roxburghiana. J. Nat. Prod. 1928, 59, 469–471. [Google Scholar] [CrossRef]
  44. Horwitz, S.B.; Chang, C.-K.; Grollman, A.P. Antiviral Action of Camptothecin. 1972. Available online: http://aac.asm.org/ (accessed on 19 March 2021).
  45. Wu, K.X.; Chu, J.J.-H. Antiviral screen identifies EV71 inhibitors and reveals camptothecin-target, DNA topoisomerase 1 as a novel EV71 host factor. Antivir. Res. 2017, 143, 122–133. [Google Scholar] [CrossRef]
  46. Wink, M. Potential of DNA Intercalating Alkaloids and Other Plant Secondary Metabolites against SARS-CoV-2 Causing COVID-19. Diversity 2020, 12, 175. [Google Scholar] [CrossRef]
  47. 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]
  48. 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] [Green Version]
  49. Valadão, A.L.C.; Abreu, C.M.; Dias, J.Z.; Arantes, P.R.; Verli, H.; Tanuri, A.; De Aguiar, R.S. Natural Plant Alkaloid (Emetine) Inhibits HIV-1 Replication by Interfering with Reverse Transcriptase Activity. Molecules 2015, 20, 11474–11489. [Google Scholar] [CrossRef] [Green Version]
  50. Bleasel, M.D.; Peterson, G.M. Emetine, Ipecac, Ipecac Alkaloids and Analogues as Potential Antiviral Agents for Coronaviruses. Pharmaceuticals 2020, 13, 51. [Google Scholar] [CrossRef] [Green Version]
  51. Mukhopadhyay, R.; Roy, S.; Venkatadri, R.; Su, Y.-P.; Ye, W.; Barnaeva, E.; Griner, L.M.; Southall, N.; Hu, X.; Wang, A.Q.; et al. Efficacy and Mechanism of Action of Low Dose Emetine against Human Cytomegalovirus. PLoS Pathog. 2016, 12, e1005717. [Google Scholar] [CrossRef] [Green Version]
  52. Choy, K.-T.; Wong, A.Y.-L.; Kaewpreedee, P.; Sia, S.F.; Chen, D.; Hui, K.P.Y.; Chu, D.K.W.; Chan, M.C.W.; Cheung, P.H.P.; Huang, X.; et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antivir. Res. 2020, 178, 104786. [Google Scholar] [CrossRef]
  53. Kim, D.E.; Min, J.S.; Jang, M.S.; Lee, J.Y.; Shin, Y.S.; Park, C.M.; Song, J.H.; Kim, H.R.; Kim, S.; Jin, Y.-H.; et al. Natural Bis-Benzylisoquinoline Alkaloids-Tetrandrine, Fangchinoline, and Cepharanthine, Inhibit Human Coronavirus OC43 Infection of MRC-5 Human Lung Cells. Biomolecules 2019, 9, 696. [Google Scholar] [CrossRef] [Green Version]
  54. Aljofan, M.; Sganga, M.L.; Lo, M.K.; Rootes, C.L.; Porotto, M.; Meyer, A.G.; Saubern, S.; Moscona, A.; Mungall, B.A. Antiviral activity of gliotoxin, gentian violet and brilliant green against Nipah and Hendra virus in vitro. Virol. J. 2009, 6, 187. [Google Scholar] [CrossRef] [Green Version]
  55. Houghton, P.J.; Woldemariam, T.Z.; Khan, A.I.; Burke, A.; Mahmood, N. Antiviral activity of natural and semi-synthetic chromone alkaloids. Antivir. Res. 1994, 25, 235–244. [Google Scholar] [CrossRef]
  56. Liu, Y.-F.; Chen, M.-H.; Guo, Q.-L.; Lin, S.; Xu, C.-B.; Jiang, Y.-P.; Li, Y.-H.; Jiang, J.-D.; Shi, J.-G. Antiviral glycosidic bisindole alkaloids from the roots ofIsatis indigotica. J. Asian Nat. Prod. Res. 2015, 17, 689–704. [Google Scholar] [CrossRef] [PubMed]
  57. 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]
  58. Chen, D.; Cai, J.; Cheng, J.; Jing, C.; Yin, J.; Jiang, J.; Peng, Z.; Hao, X. Design, Synthesis and Structure-Activity Relationship Optimization of Lycorine Derivatives for HCV Inhibition. Sci Rep. 2015, 5, 14972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. He, J.; Qi, W.-B.; Wang, L.; Tian, J.; Jiao, P.; Liu, G.-Q.; Ye, W.-C.; Liao, M. Amaryllidaceae alkaloids inhibit nuclear-to-cytoplasmic export of ribonucleoprotein (RNP) complex of highly pathogenic avian influenza virus H5N1. Influ. Other Respir. Viruses 2013, 7, 922–931. [Google Scholar] [CrossRef] [Green Version]
  60. Wang, P.; Li, L.; Wang, Q.-Y.; Shang, L.-Q.; Shi, P.-Y.; Yin, Z. Anti-Dengue-Virus Activity and Structure-Activity Relationship Studies of Lycorine Derivatives. ChemMedChem 2014, 9, 1522–1533. [Google Scholar] [CrossRef]
  61. Li, S.-Y.; Chen, C.; Zhang, H.-Q.; Guo, H.-Y.; Wang, H.; Wang, L.; Zhang, X.; Hua, S.-N.; Yu, J.; Xiao, P.-G.; et al. Identification of natural compounds with antiviral activities against SARS-associated coronavirus. Antivir. Res. 2005, 67, 18–23. [Google Scholar] [CrossRef]
  62. Palem, J.; Bedadala, G.; El Sayed, K.; Hsia, S.-C. Manzamine A as a Novel Inhibitor of Herpes Simplex Virus Type-1 Replication in Cultured Corneal Cells. Planta Med. 2010, 77, 46–51. [Google Scholar] [CrossRef] [Green Version]
  63. McMahon, J.B.; Currens, M.J.; Gulakowski, R.J.; Buckheit, R.W., Jr.; Lackman-Smith, C.; Hallock, Y.F.; Boyd, M.R. Michellamine B, a Novel Plant Alkaloid, Inhibits Human Immunodeficiency Virus-Induced Cell Killing by at Least Two Distinct Mechanisms. Antimicrob. Agents Chemother. 1995, 39, 484–488. [Google Scholar] [CrossRef] [Green Version]
  64. Boyd, M.R.; Hallock, Y.F.; Cardellina, J.H.; Manfredi, K.P.; Blunt, J.W.; McMahon, J.B.; Buckheit, R.W., Jr.; Bringmann, G.; Schäffer, M.; Cragg, G.M.; et al. Anti-HIV Michellamines from Ancistrocladus korupensisl. J. Med. Chem. 1994, 37, 1740–1745. [Google Scholar] [CrossRef]
  65. Montanha, J.A.; Amoros, M.; Boustie, J.; Girre, L. Anti-Herpes Virus Activity of Aporphine Alkaloids. Planta Med. 1995, 61, 419–424. [Google Scholar] [CrossRef] [PubMed]
  66. Loya, S.; Rudi, A.; Kashman, Y.; Hizi, A. Polycitone A, a novel and potent general inhibitor of retroviral reverse transcriptases and cellular DNA polymerases. Biochem. J. 1999, 344, 85–92. [Google Scholar] [CrossRef] [PubMed]
  67. Wu, C.-Y.; Jan, J.-T.; Ma, S.-H.; Kuo, C.-J.; Juan, H.-F.; Cheng, Y.-S.E.; Hsu, H.-H.; Huang, H.-C.; Wu, D.; Brik, A.; et al. Small molecules targeting severe acute respiratory syndrome human coronavirus. Proc. Natl. Acad. Sci. USA 2004, 101, 10012–10017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Serkedjieva, J.; Velcheva, M. In Vitro Anti-Influenza Virus Activity of the Pavine Alkaloid (-)-Thalimonine Isolated from Thalictrum Simplex L. Antivir. Chem. Chemother. 2003, 14, 75–80. [Google Scholar] [CrossRef] [Green Version]
  69. Troost, B.; Mulder, L.M.; Diosa-Toro, M.; Van De Pol, D.; Rodenhuis-Zybert, I.A.; Smit, J.M. Tomatidine, a natural steroidal alkaloid shows antiviral activity towards chikungunya virus in vitro. Sci. Rep. 2020, 10, 6364. [Google Scholar] [CrossRef] [Green Version]
  70. Diosa-Toro, M.; Troost, B.; van de Pol, D.; Heberle, A.M.; Urcuqui-Inchima, S.; Thedieck, K.; Smit, J.M. Tomatidine, a novel antiviral compound towards dengue virus. Antivir. Res. 2019, 161, 90–99. [Google Scholar] [CrossRef]
  71. Silva, E.M.; Cirne-Santos, C.C.; Frugulhetti, I.C.P.P.; Galvão-Castro, B.; Saraiva, E.M.B.; Kuehne, M.E.; Bou-Habib, D.C. Anti-HIV-1 Activity of the Iboga Alkaloid Congener 18-Methoxycoronaridine. Planta Med. 2004, 70, 808–812. [Google Scholar] [CrossRef]
  72. Nisar, T.; Wang, Z.-C.; Yang, X.; Tian, Y.; Iqbal, M.; Guo, Y. Characterization of citrus pectin films integrated with clove bud essential oil: Physical, thermal, barrier, antioxidant and antibacterial properties. Int. J. Biol. Macromol. 2018, 106, 670–680. [Google Scholar] [CrossRef]
  73. Zhou, Y.; Simmons, G. Development of novel entry inhibitors targeting emerging viruses. Expert Rev. Anti-Infect. Ther. 2012, 10, 1129–1138. [Google Scholar] [CrossRef]
  74. Cohen, S.; Au, S.; Panté, N. How viruses access the nucleus. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2011, 1813, 1634–1645. [Google Scholar] [CrossRef] [Green Version]
  75. Dimitrov, D.S. Virus entry: Molecular mechanisms and biomedical applications. Nat. Rev. Genet. 2004, 2, 109–122. [Google Scholar] [CrossRef] [PubMed]
  76. Lingwood, C.A. Glycosphingolipid Functions. Cold Spring Harb. Perspect. Biol. 2011, 3, a011874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wolf, M.C.; Freiberg, A.N.; Zhang, T.; Akyol-Ataman, Z.; Grock, A.; Hong, P.W.; Li, J.; Watson, N.F.; Fang, A.Q.; Aguilar, H.C.; et al. A broad-spectrum antiviral targeting entry of enveloped viruses. Proc. Natl. Acad. Sci. USA 2010, 107, 3157–3162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Dai, L.; Song, J.; Lu, X.; Deng, Y.-Q.; Musyoki, A.M.; Cheng, H.; Zhang, Y.; Yuan, Y.; Song, H.; Haywood, J.; et al. Structures of the Zika Virus Envelope Protein and Its Complex with a Flavivirus Broadly Protective Antibody. Cell Host Microbe 2016, 19, 696–704. [Google Scholar] [CrossRef] [PubMed]
  79. Carette, J.E.; Raaben, M.; Wong, A.C.; Herbert, A.S.; Obernosterer, G.; Mulherkar, N.; Kuehne, A.I.; Kranzusch, P.J.; Griffin, A.M.; Ruthel, G.; et al. Ebola virus entry requires the cholesterol transporter Niemann–Pick C1. Nature 2011, 477, 340–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Hung, T.-C.; Jassey, A.; Liu, C.-H.; Lin, C.-J.; Lin, C.-C.; Wong, S.H.; Wang, J.Y.; Yen, M.-H.; Lin, L.-T. Berberine inhibits hepatitis C virus entry by targeting the viral E2 glycoprotein. Phytomedicine 2018, 53, 62–69. [Google Scholar] [CrossRef]
  81. Xia, S.; Zhu, Y.; Liu, M.; Lan, Q.; Xu, W.; Wu, Y.; Ying, T.; Liu, S.; Shi, Z.; Jiang, S.; et al. Fusion mechanism of 2019-nCoV and fusion inhibitors targeting HR1 domain in spike protein. Cell. Mol. Immunol. 2020, 17, 765–767. [Google Scholar] [CrossRef]
  82. Maurya, V.K.; Kumar, S.; Prasad, A.K.; Bhatt, M.L.B.; Saxena, S.K. Structure-based drug designing for potential antiviral activity of selected natural products from Ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. VirusDisease 2020, 31, 179–193. [Google Scholar] [CrossRef]
  83. Fielding, B.C.; Filho, C.D.S.M.B.; Ismail, N.S.M.; De Sousa, D.P. Alkaloids: Therapeutic Potential against Human Coronaviruses. Molecules 2020, 25, 5496. [Google Scholar] [CrossRef]
  84. Gyebi, G.A.; Adegunloye, A.P.; Ibrahim, I.M.; Ogunyemi, O.M.; Afolabi, S.O.; Ogunro, O.B. Prevention of SARS-CoV-2 cell entry: Insight from in silico interaction of drug-like alkaloids with spike glycoprotein, human ACE2, and TMPRSS2. J. Biomol. Struct. Dyn. 2020, 1–25. [Google Scholar] [CrossRef]
  85. Lebecque, S.; Crowet, J.-M.; Lins, L.; Delory, B.M.; du Jardin, P.; Fauconnier, M.-L.; Deleu, M. Interaction between the barley allelochemical compounds gramine and hordenine and artificial lipid bilayers mimicking the plant plasma membrane. Sci. Rep. 2018, 8, 9784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lundbæk, J.A.; Birn, P.; Tape, S.E.; Toombes, G.E.S.; Søgaard, R.; Koeppe, R.; Gruner, S.M.; Hansen, A.J.; Andersen, O.S. Capsaicin Regulates Voltage-Dependent Sodium Channels by Altering Lipid Bilayer Elasticity. Mol. Pharmacol. 2005, 68, 680–689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Torrecillas-Sanchez, A.; Schneider, M.; Fernández-Martínez, A.M.; Ausili, A.; De Godos, A.M.; Corbalan, S.; Gomez-Fernandez, J.C. Capsaicin Fluidifies the Membrane and Localizes Itself near the Lipid–Water Interface. ACS Chem. Neurosci. 2015, 6, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
  88. Wink, M.; Schmeller, T.; Latz-Brüning, B. Modes of Action of Allelochemical Alkaloids: Interaction with Neuroreceptors, DNA, and Other Molecular Targets. J. Chem. Ecol. 1998, 24, 1881–1937. [Google Scholar] [CrossRef]
  89. Zidovetzki, R.; Sherman, I.W.; Atiya, A.; De Boeck, H. A nuclear magnetic resonance study of the interactions of the antimalarials chloroquine, quinacrine, quinine and mefloquine with dipalmitoylphosphatidylcholine bilayers. Mol. Biochem. Parasitol. 1989, 35, 199–207. [Google Scholar] [CrossRef]
  90. Chen, X.; Si, L.; Liu, D.; Proksch, P.; Zhang, L.; Zhou, D.; Lin, W. Neoechinulin B and its analogues as potential entry inhibitors of influenza viruses, targeting viral hemagglutinin. Eur. J. Med. Chem. 2015, 93, 182–195. [Google Scholar] [CrossRef]
  91. Ho, Y.-J.; Lu, J.-W.; Huang, Y.-L.; Lai, Z.-Z. Palmatine inhibits Zika virus infection by disrupting virus binding, entry, and stability. Biochem. Biophys. Res. Commun. 2019, 518, 732–738. [Google Scholar] [CrossRef]
  92. Matsuda, K.; Hattori, S.; Komizu, Y.; Kariya, R.; Ueoka, R.; Okada, S. Cepharanthine inhibited HIV-1 cell–cell transmission and cell-free infection via modification of cell membrane fluidity. Bioorganic Med. Chem. Lett. 2014, 24, 2115–2117. [Google Scholar] [CrossRef]
  93. Shao, J.; Zeng, D.; Tian, S.; Liu, G.; Fu, J. Identification of the natural product berberine as an antiviral drug. AMB Express 2020, 10, 1–11. [Google Scholar] [CrossRef]
  94. 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] [Green Version]
  95. Basu, A.; Kumar, G.S. Nucleic acids binding strategies of small molecules: Lessons from alkaloids. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2018, 1862, 1995–2016. [Google Scholar] [CrossRef] [PubMed]
  96. Walsh, D.; Mohr, I. Viral subversion of the host protein synthesis machinery. Nat. Rev. Microbiol. 2011, 9, 860–875. [Google Scholar] [CrossRef] [PubMed]
  97. Said, Z.; Abdelwahab, K. Antiviral replication agents. In Viral Replication; Rosas-Acosta, G., Ed.; InTech Open Science: London, UK, 2013; p. 144. [Google Scholar]
  98. Payne, S. Introduction to RNA Viruses. Viruses 2017, 97–105. [Google Scholar] [CrossRef]
  99. Rampersad, S.; Tennant, P. Replication and Expression Strategies of Viruses. Viruses 2018, 55–82. [Google Scholar] [CrossRef]
  100. Esposito, F.; Corona, A.; Tramontano, E. HIV-1 Reverse Transcriptase Still Remains a New Drug Target: Structure, Function, Classical Inhibitors, and New Inhibitors with Innovative Mechanisms of Actions. Mol. Biol. Int. 2012, 2012, 586401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Chen, H.; Beardsley, G.P.; Coen, D.M. Mechanism of ganciclovir-induced chain termination revealed by resistant viral polymerase mutants with reduced exonuclease activity. Proc. Natl. Acad. Sci. USA 2014, 111, 17462–17467. [Google Scholar] [CrossRef] [Green Version]
  102. Dial, C.N.; Tate, P.M.; Kicmal, T.M.; Mounce, B.C. Coxsackievirus B3 Responds to Polyamine Depletion via Enhancement of 2A and 3C Protease Activity. Viruses 2019, 11, 403. [Google Scholar] [CrossRef] [Green Version]
  103. Mounce, B.C.; Cesaro, T.; Moratorio, G.; Hooikaas, P.J.; Yakovleva, A.; Werneke, S.W.; Smith, E.C.; Poirier, E.Z.; Simon-Loriere, E.; Prot, M.; et al. Inhibition of Polyamine Biosynthesis Is a Broad-Spectrum Strategy against RNA Viruses. J. Virol. 2016, 90, 9683–9692. [Google Scholar] [CrossRef] [Green Version]
  104. Chang, K.-O.; Kim, Y.; Lovell, S.; Rathnayake, A.D.; Groutas, W.C. Antiviral Drug Discovery: Norovirus Proteases and Development of Inhibitors. Viruses 2019, 11, 197. [Google Scholar] [CrossRef] [Green Version]
  105. Zhu, W.; Chen, C.Z.; Gorshkov, K.; Xu, M.; Lo, D.C.; Zheng, W. RNA-Dependent RNA Polymerase as a Target for COVID-19 Drug Discovery. SLAS Discov. Adv. Sci. Drug Discov. 2020, 25, 1141–1151. [Google Scholar] [CrossRef]
  106. Almaqwashi, A.A.; Paramanathan, T.; Rouzina, I.; Williams, M.C. Mechanisms of small molecule–DNA interactions probed by single-molecule force spectroscopy. Nucleic Acids Res. 2016, 44, 3971–3988. [Google Scholar] [CrossRef] [PubMed]
  107. Palchaudhuri, R.; Hergenrother, P.J. DNA as a target for anticancer compounds: Methods to determine the mode of binding and the mechanism of action. Curr. Opin. Biotechnol. 2007, 18, 497–503. [Google Scholar] [CrossRef] [PubMed]
  108. Woo, S.H.; Sun, N.-J.; Cassady, J.M.; Snapka, R.M. Topoisomerase II inhibition by aporphine alkaloids. Biochem. Pharmacol. 1999, 57, 1141–1145. [Google Scholar] [CrossRef]
  109. Krstin, S.; Mohamed, T.; Wang, X.; Wink, M. How do the alkaloids emetine and homoharringtonine kill trypanosomes? An insight into their molecular modes of action. Phytomedicine 2016, 23, 1771–1777. [Google Scholar] [CrossRef]
  110. Sinha, R.; Kumar, G.S. Interaction of Isoquinoline Alkaloids with an RNA Triplex: Structural and Thermodynamic Studies of Berberine, Palmatine, and Coralyne Binding to Poly(U).Poly(A)*Poly(U). J. Phys. Chem. B 2009, 113, 13410–13420. [Google Scholar] [CrossRef]
  111. Schmeller, T.; Latz-Brüning, B.; Wink, M. Biochemical activities of berberine, palmatine and sanguinarine mediating chemical defence against microorganisms and herbivores. Phytochemistry 1997, 44, 257–266. [Google Scholar] [CrossRef]
  112. Sethi, M.L. Enzyme Inhibition VIII: Mode of Inhibition of Reverse Transcriptase Activity by Analogues, Isomers, and Related Alkaloids of Coralyne. J. Pharm. Sci. 1985, 74, 889–891. [Google Scholar] [CrossRef]
  113. Low, J.S.Y.; Chen, K.C.; Wu, K.X.; Ng, M.M.-L.; Chu, J.J.H. Antiviral Activity of Emetine Dihydrochloride Against Dengue Virus Infection. J. Antivir. Antiretrovir. 2009, 1, 62–71. [Google Scholar] [CrossRef] [Green Version]
  114. Contrerast, A.; Carrascot, L. Selective Inhibition of Protein Synthesis in Virus-Infected Mammalian Cells. 1979. Available online: http://jvi.asm.org/ (accessed on 7 August 2021).
  115. Grollman, A.P. No Tinhibitor of protein biosynthesisitle. J. Biol. Chem. 1968, 245, 4089–4094. [Google Scholar] [CrossRef]
  116. Kantarjian, H.M.; O’Brien, S.; Cortes, J. Homoharringtonine/Omacetaxine Mepesuccinate: The Long and Winding Road to Food and Drug Administration Approval. Clin. Lymphoma Myeloma Leuk. 2013, 13, 530–533. [Google Scholar] [CrossRef] [Green Version]
  117. Gupta, R.S.; Siminovitch, L. The molecular basis of emetine resistance in chinese hamster ovary cells: Alteration in the 40S ribosomal subunit. Cell 1977, 10, 61–66. [Google Scholar] [CrossRef]
  118. Jimenez, A.; Carrasco, L.; Vazquez, D. Enzymic and Nonenzymic Translocation by Yeast Polysomes. Site of Action of a Number of Inhibitors? Biochemistry 1977, 16, 4727–4730. [Google Scholar] [CrossRef] [PubMed]
  119. Fresno, M.; Jimenez, A.; Vazquez, D. Inhibition of Translation in Eukaryotic Systems by Harringtonine. Eur. J. Biochem. 1977, 72, 323–330. [Google Scholar] [CrossRef] [PubMed]
  120. Ramabhadran, T.V.; Thach, R.E. Specificity of protein synthesis inhibitors in the inhibition of encephalomyocarditis virus replication. J. Virol. 1980, 34, 293–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Liu, J.; Yang, Y.; Xu, Y.; Ma, C.; Qin, C.; Zhang, L. Lycorine reduces mortality of human enterovirus 71-infected mice by inhibiting virus replication. Virol. J. 2011, 8, 483–483. [Google Scholar] [CrossRef] [Green Version]
  122. Jimenez, A.; Santos, A.; Alonso, G.; Vazquez, D. Inhibitors of protein synthesis in eukaryotic cells: Comparative effects of some Amaryllidaceae alkaloids. Biochim. Biophys. Acta (BBA)-Nucleic Acids Protein Synth. 1976, 425, 342–348. [Google Scholar] [CrossRef]
  123. Kukhanova, M.; Victorova, L.; Krayevsky, A. Peptidyltransferase center of ribosomes: On the mechanism of action of alkaloid lycorine. FEBS Lett. 1983, 160, 129–133. [Google Scholar] [CrossRef] [Green Version]
  124. Baroni, A.; Paoletti, I.; Ruocco, E.; Ayala, F.; Corrado, F.; Wolf, R.; Tufano, M.A.; Donnarumma, G. Antiviral effects of quinine sulfate on HSV-1 HaCat cells infected: Analysis of the molecular mechanisms involved. J. Dermatol. Sci. 2007, 47, 253–255. [Google Scholar] [CrossRef]
  125. Malakar, S.; Sreelatha, L.; Dechtawewat, T.; Noisakran, S.; Yenchitsomanus, P.T.; Chu, J.J.H.; Limjindaporn, T. Drug repurposing of quinine as antiviral against dengue virus infection. Virus Res. 2018, 255, 171–178. [Google Scholar] [CrossRef]
  126. Moertel, C.G.; Schutt, A.J.; Reitemeier, R.J.; Hahn, R.G. Phase II study of camptothecin (NSC-100880) in the treatment of advanced gastrointestinal cancer. Cancer Chemother. Rep. 1972, 65, 95–101. [Google Scholar]
  127. Bennett, R.P.; Stewart, R.A.; Hogan, P.A.; Ptak, R.G.; Mankowski, M.K.; Hartman, T.L.; Buckheit, R.W.; Snyder, B.A.; Salter, J.D.; Morales, G.; et al. An analog of camptothecin inactive against Topoisomerase I is broadly neutralizing of HIV-1 through inhibition of Vif-dependent APOBEC3G degradation. Antivir. Res. 2016, 136, 51–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Cao, Z.; Kozielski, A.; Liu, X.; Wang, Y.; Vardeman, D.; Giovanella, B. Crystalline Camptothecin-20( S )-O-Propionate Hydrate: A Novel Anticancer Agent with Strong Activity against 19 Human Tumor Xenografts. Cancer Res. 2009, 69, 4742–4749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Clinicaltrials.gov. Phase I Clinical Trial of Camptothecin-20-O-Propionate Hydrate (CZ48). Available online: https://clinicaltrials.gov/ct2/show/study/NCT02575638 (accessed on 22 October 2021).
  130. Lancelot, A.; Clavería-Gimeno, R.; Velazquez-Campoy, A.; Abian, O.; Serrano, J.L.; Sierra, T. Nanostructures based on ammonium-terminated amphiphilic Janus dendrimers as camptothecin carriers with antiviral activity. Eur. Polym. J. 2017, 90, 136–1497. [Google Scholar] [CrossRef]
  131. Garner-Wizard, M.; Henson, S.; Milot, B.; Minigh, J.; Oliff, H.S.; Oppel, M. Update on Plant-Derived Compounds Used on HIV Treatment. 2009. Available online: https://www.herbalgram.org/media/4849/373_review110685-373-110685.pdf (accessed on 25 October 2021).
  132. Cragg, G.M.; Newman, D.J.; Kingston, D.G.I. Terrestrial plants as a source of novel pharmaceutical agents. Compr. Nat. Prod. II Chem. Biol. 2010, 2, 5–39. [Google Scholar] [CrossRef]
  133. Lu, Y. The Design and Synthesis of Novel Michellamine B Analogues Targeting HIV; University of Wollongong: Wollongong, Australia, 2015. [Google Scholar]
  134. Wishart, D.S.; Knox, C.; Guo, A.C.; Shrivastava, S.; Hassanali, M.; Stothard, P.; Chang, Z.; Woolsey, J. DrugBank: A comprehensive resource for in silico drug discovery and exploration. Nucleic Acids Res. 2006, 34, D668–D672. [Google Scholar] [CrossRef]
  135. Nadia Bouabdallaoui, F.; Tardif, J.C. The Role of Colchicine in Recent Clinical Trials—A Focused Review on Pericardial Disease. American College of Cardiology. 2020. Available online: https://www.acc.org/latest-in-cardiology/articles/2020/08/13/13/11/the-role-of-colchicine-in-recent-clinical-trials (accessed on 29 October 2021).
  136. Deftereos, S.G.; Giannopoulos, G.; Vrachatis, D.A.; Siasos, G.D.; Giotaki, S.G.; Gargalianos, P.; Metallidis, S.; Sianos, G.; Baltagiannis, S.; Panagopoulos, P.; et al. Effect of Colchicine vs Standard Care on Cardiac and Inflammatory Biomarkers and Clinical Outcomes in Patients Hospitalized With Coronavirus Disease 2019: The GRECCO-19 randomized clinical trial. JAMA Netw. Open 2020, 3, e2013136. [Google Scholar] [CrossRef]
  137. Lopes, M.I.; Bonjorno, L.P.; Giannini, M.C.; Amaral, N.B.; Menezes, P.I.; Dib, S.M.; Gigante, S.L.; Benatti, M.N.; Rezek, U.C.; Emrich-Filho, L.L.; et al. Beneficial effects of colchicine for moderate to severe COVID-19: A randomised, double-blinded, placebo-controlled clinical trial. RMD Open 2021, 7, e001455. [Google Scholar] [CrossRef]
  138. Rubiaceae, S. Ipecacuanha, emetine, and dehydroemetine. Meyler’s Side Eff. Drugs 2016, 311–312. [Google Scholar] [CrossRef]
  139. Zbinden, G.; Kleinert, R.; Rageth, B. Assessment of Emetine Cardiotoxicity in a Subacute Toxicity Experiment in Rats. J. Cardiovasc. Pharmacol. 1980, 2, 155–164. [Google Scholar] [CrossRef]
  140. Matthews, H.; Deakin, J.; Rajab, M.; Idris-Usman, M.; Nirmalan, N.J. Investigating antimalarial drug interactions of emetine dihydrochloride hydrate using CalcuSyn-based interactivity calculations. PLoS ONE 2017, 12, e0173303. [Google Scholar] [CrossRef] [Green Version]
  141. Kretzing, S.; Abraham, G.; Seiwert, B.; Ungemach, F.R.; Krügel, U.; Regenthal, R. Dose-dependent emetic effects of the Amaryllidaceous alkaloid lycorine in beagle dogs. Toxicon 2011, 57, 117–124. [Google Scholar] [CrossRef] [PubMed]
  142. Roy, M.; Liang, L.; Xiao, X.; Feng, P.; Ye, M.; Liu, J. Lycorine: A prospective natural lead for anticancer drug discovery. Biomed. Pharmacother. 2018, 107, 615–624. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, P.; Yuan, X.; Yu, T.; Huang, H.; Yang, C.; Zhang, L.; Yang, S.; Luo, X.; Luo, J. Lycorine inhibits cell proliferation, migration and invasion, and primarily exerts in vitro cytostatic effects in human colorectal cancer via activating the ROS/p38 and AKT signaling pathways. Oncol. Rep. 2021, 45, 19. [Google Scholar] [CrossRef] [PubMed]
  144. Cortes, J.; Lipton, J.H.; Rea, D.; Digumarti, R.; Chuah, C.; Nanda, N.; Benichou, A.-C.; Craig, A.R.; Michallet, M.; Nicolini, F.E.; et al. Phase 2 study of subcutaneous omacetaxine mepesuccinate after TKI failure in patients with chronic-phase CML with T315I mutation. Blood 2012, 120, 2573–2580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Nicolini, F.E.; Khoury, H.J.; Akard, L.; Rea, D.; Kantarjian, H.; Baccarani, M.; Leonoudakis, J.; Craig, A.; Benichou, A.C.; Cortes, J. Omacetaxine mepesuccinate for patients with accelerated phase chronic myeloid leukemia with resistance or intolerance to two or more tyrosine kinase inhibitors. Haematologica 2013, 98, 78–79. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Chemical structure of alkaloids.
Figure 1. Chemical structure of alkaloids.
Molecules 27 00903 g001aMolecules 27 00903 g001bMolecules 27 00903 g001c
Table 1. Antiviral activities of alkaloids.
Table 1. Antiviral activities of alkaloids.
CompoundSourceVirusDose
IC50 or EC50
ActivityExperimentRef
AnisomycinPureDENV,
ZIKAV
7.6 nM
15.9 nM
Inhibits replicationIn vitro/In vivo[39]
BerberineBerberis vulgaris (Berberidaceae)HCMV0.68 μMInhibits the virus replication
cycle between viral attachment/entry and genomic DNA replication
In vitro[40]
HSV-1 and 26.77 and 5.04 μM[41]
Hydrastis CanadensisH1N10.01 μMInhibited production of proteins[42]
Buchapine,Euodia roxburghianaHIV-10.94 μMInhibit reverse transcriptaseIn vitro[43]
Ophiorrhiza mungosVACV10 μM,Inhibition of viral DNA synthesisIn vitro[44]
CamptothecinCamptotheca acuminataEV7110 μMInhibits viral RNA replication and translationIn vitro,[45]
ColchicineHaplophyllum tuberculatum, Colchicum autumnaleHIV 11–10Inhibit replication by DNA intercalationIn vitro[46]
EmetineCephaelis ipecacuanha A.RNA, DNA viruses3.03 ngInhibition of viral protein synthesisIn vitro/In vivo[47]
ZIKAV, EVD10 μMInhibition of ZIKAV NS5 polymerase activity and disruption of lysosomal function,
inhibits EBOV entry andreplication of DNA viruses
In vitro/In vivo[48]
HIV0.72 μMInhibits HIV-1 Replication by interfering with reverse transcriptase activityIn vitro[49]
SARS, MERS 0.0135 μMReplication inhibitionIn vitro[50]
HCMV0.0087–0.04 μMInhibition on ribosomal processing S14 (RPS14) bindingIn vitro[51]
SARS-CoV-20.46 μMReplication inhibitionIn vitro[52]
FangchinolinStephania tetrandraHCV10 µMSuppressed the replication and inhibited viral S and N protein expressionIn vitro[53]
GliotoxinPureHI10 μMInhibits intracellular replicationIn vitro[54]
Hemanthamene H5N14·15 μMInhibits the translocation of the ribonucleoprotein complexIn vitro[55]
HomorringtonineCephalotoxus fortuneSARS-CoV-22.55 μMReplication inhibitionIn vitro[52]
Isatindigobisindoloside F.Isatis indigoticaCVB38.4 μMInhibit the replicationIn vitro[56]
LycorineClivia miniateZIKAV0.01 to 10 μMInhibits viral replication by restraining RdRp activityIn vitro/In vivo[57]
HCV6.10 μMInhibits viral replication by the expression of hsc70 in host cellsIn vitro[58]
H5N10·52 μMInhibits the translocation of the ribonucleoprotein complexIn vitro[59]
DENV0.4 μMSuppression of viral RNA replicationIn vitro[60]
L.radiataSARS-CoV15.7 nMViral inhibitionIn vitro[61]
ManzamineAcanthostrongylophoraHSV-11 µMRepressed ICP0 transcriptionIn vitro[62]
Michellamines BA. korupensisHIV1 μMInhibiting cellular fusion and syncytium formationIn vitro[63]
Michellamines A, B, and CAncistrocladus korupensisHIV1, HIV22–10 μMComplete inhibition of the replication and cytopathic effects of HIVIn vitro/In vivo[64]
OliverinePolyathia oliveri;HSV-17.5 μMInhibition of viral DNA synthesisIn vitro[65]
Polycitone APolycitor spHIV, MLV, MMTV295 nMPotent inhibitory activity on both RNA- and DNA-directed DNA polymerasesIn vitro[66]
ReserpinePureSARS3.4 μMInhibits 3CL protease and viral entryIn vitro[67]
SchumannificineSchumanniophyton magnificum;HSV1.6 μMIrreversible binding to gp120In vitro[53]
Tetrandrine,Stephania tetrandraHCV10 µMSuppressed the replication and inhibited viral S and N protein expressionIn vitro[53]
ThalimonineThalictrum simplexH7N70.1 µMInhibits viral reproductionIn vitro[68]
TomatidineSkin and leaves of tomatoesCHIKV1.3 µMInhibits infection of three different CHIKV genotypesIn vitro[69]
DENV0.82 μMInhibit replicationIn vitro[70]
18-methoxycoronaidineTabernanthe ibogaHIV-19.5 μMInhibit the replication of primary isolates of HIVIn vitro[71]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abookleesh, F.L.; Al-Anzi, B.S.; Ullah, A. Potential Antiviral Action of Alkaloids. Molecules 2022, 27, 903. https://doi.org/10.3390/molecules27030903

AMA Style

Abookleesh FL, Al-Anzi BS, Ullah A. Potential Antiviral Action of Alkaloids. Molecules. 2022; 27(3):903. https://doi.org/10.3390/molecules27030903

Chicago/Turabian Style

Abookleesh, Frage L., Bader S. Al-Anzi, and Aman Ullah. 2022. "Potential Antiviral Action of Alkaloids" Molecules 27, no. 3: 903. https://doi.org/10.3390/molecules27030903

Article Metrics

Back to TopTop