Planta Med 2022; 88(12): 1047-1059
DOI: 10.1055/a-1581-3707
Biological and Pharmacological Activity
Original Papers

Activity of THC, CBD, and CBN on Human ACE2 and SARS-CoV1/2 Main Protease to Understand Antiviral Defense Mechanism

1   Technical Biochemistry, Faculty of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
,
1   Technical Biochemistry, Faculty of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
2   MINDbioscience GmbH, Dortmund, Germany
,
1   Technical Biochemistry, Faculty of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
› Author Affiliations

Supported by: Deutscher Akademischer Austauschdienst 57299294
 

Abstract

THC, CBD, and CBN were reported as promising candidates against SARS-CoV2 infection, but the mechanism of action of these three cannabinoids is not understood. This study aims to determine the mechanism of action of THC, CBD, and CBN by selecting two essential targets that directly affect the coronavirus infections as viral main proteases and human angiotensin-converting enzyme2. Tested THC and CBD presented a dual-action action against both selected targets. Only CBD acted as a potent viral main protease inhibitor at the IC50 value of 1.86 ± 0.04 µM and exhibited only moderate activity against human angiotensin-converting enzyme2 at the IC50 value of 14.65 ± 0.47 µM. THC acted as a moderate inhibitor against both viral main protease and human angiotensin-converting enzymes2 at the IC50 value of 16.23 ± 1.71 µM and 11.47 ± 3.60 µM, respectively. Here, we discuss cannabinoid-associated antiviral activity mechanisms based on in silico docking studies and in vitro receptor binding studies.


#

Abbreviations

CBD: Cannabidiol
CBN: Cannabinol
RMSD: Root-mean-square deviation of atomic positions
SARS-CoV2: Severe acute respiratory syndrome coronavirus2
THC: Δ-9-Tetrahydrocannabinol
 

Introduction

Plant secondary metabolites have been intensively studied for novel antiviral substances long before the severe acute respiratory syndrome coronavirus2 or SARS-CoV2 pandemic occurred [1], [2], [3], [4], but today they gain more attention than before [5], [6], [7]. The principle of antiviral drug discovery from plants is that plants developed defensive metabolites against pathogenic microorganisms such as bacteria, fungi, and viruses to survive [8], [9]. A success story of an approved plant metabolite against the human viral infection is podophyllotoxin, a lignan from Podophyllum peltatum L. or Podophyllum emodi ROYLE, approved to treat human papillomavirus infection topically [9], [10]. However, there are much more plant metabolites that are currently under investigation [11]. Three examples will be given here. First is homoharringtonine, a plant alkaloid, from Cephalotaxus harringtonii (Knight ex J.Forbes) K.Koch [12], [13] showing a wide range of antiviral activities against hepatitis, herpes simplex1, varicella-zoster, foot and mouse disease, Ebola, and SARS-CoV2 viruses [11], [14], [15], [16], [17]. Another example is emetine (a tetrahydroisoquinoline alkaloid) from the plant Psychotria ipecacuanha (Brot.) Stoves [18], [19]. Emetine also exhibits a wide range of antiviral activities against hepatitis, Zika, Ebola, Middle-East respiratory syndrome coronavirus or MERS-CoV, SARS-CoV1, and SARS-CoV2 viruses [11], [17], [18], [20], [21]. Ribaudo et al. (2021) reported that artemisinin (a known antimalarial drug) blocked the entry of SARS-CoV2 into human cells by inhibiting human angiotensin-converting enzyme2 (hACE2) [22].

Since the uprise of this pandemic, the interest in cannabinoids for the SARS-CoV2 treatment has been increased remarkably, but the direct antiviral activity of cannabinoids is still low [23]. Most of the studies were review article focusing on the supportive therapy for SARS-CoV2 infection such as lowering the inflammation by reducing the cytokine storm, especially interleukin-6 and the agonistic effect of the cannabinoids at PPARγ receptor at lung tissue [24], [25], [26], [27]. Only a few studies have reported the activity of cannabinoids against SARS-CoV2 infection experimentally. One study from Wang et al. (2020) showed the downregulation of hACE2 from CBD extract high content [28]. Another study is the first and only study from Raj et al. (2021) that reported the direct and remarkably activity of THC, CBD, and CBN against SARS-CoV2 infection with the same potency as the standard drugs such as chloroquine, remdesivir, and lopinavir in the cell-based assay [29]. However, the mechanism of action of these three cannabinoids against SARS-CoV2 infection is not yet known for certain. Therefore, this study aims to reveal the antiviral mechanism of THC, CBD, and CBN.

Based on the coronavirus pathogenesis (both SARS-CoV1 and SARS-CoV2), there are two main strategies to stop these coronaviruses infection. The first strategy is to prevent viruses from entering the cell. The receptor protein hACE2, is located on the membrane of immune cells, is considered essential for cell invasion, and the viral spike protein, a transmembrane protease, serine 2 (or TRPRSS2), is critical for entering the cell. The second strategy is to stop replicating the virus in the host cell by inhibiting the viral proteolysis (stopping the viral main protease activity). Therefore, we selected hACE2 and viral main proteases from both SARS-CoV1 and SARS-CoV2 as the target proteins to determine the mechanism of action and evaluate the potency of THC, CBD, and CBN against coronavirus infections, as presented in [Fig. 1].

Zoom Image
Fig. 1 Simplified replication cycle of coronaviruses in the human host cell: (1) indicates one of our targets, inhibiting hACE2 activity in which will prevent the coronaviruses to enter the human cell; (2) indicates the other target, inhibiting the viral main protease in which will stop the viral replication.

#

Results

The antiviral activity of THC, CBD, and CBN against SARS-CoV2 infection was recently reported from a cell-based assay, but still the mechanism of action of mentioned cannabinoids has not yet been elucidated [29]. Currently, only the predicted mechanism was proposed by a computational experiment [29], [30]. Our main objective was to evaluate and confirm the antiviral action of three cannabinoids against coronavirus main proteases by tracking the fluorogenic substrate to monitor the enzyme activity. As shown in [Fig. 2], only THC and CBD have strong inhibitory activity against SARS-CoV1 and SARS-CoV2 main proteases at the screening concentration of 100 µg/mL with a minor difference from the positive standard GC376 (Fig. S7, Supporting Information). [Fig. 2] demonstrates high inhibitory activity (IC50) of THC and CBD at 104.43 ± 6.27% and 100.78 ± 1.10% for SARS-CoV1 main proteases, 100.00 ± 6.15% and 102.12 ± 1.66% for SARS-CoV2 main protease, respectively. Remarkably, inhibitions were superior to the activity of GC376 as reference (88.15 ± 19.41%) against SARS-CoV1 main protease and were equal to SARS-CoV2 main protease at 100.62 ± 4.80%, respectively. CBN showed only a minor effect against SARS-CoV1 and SARS-CoV2 main proteases (39.59 ± 10.79% and 31.46 ± 18.49%, respectively). Data support the proposed mechanism from an earlier report suggesting that all three cannabinoids (THC, CBD, and CBN) inhibit coronavirus main protease activity [29]. Since both THC and CBD were characterized as positive candidates (% inhibition higher than 80%), both cannabinoids were subjected to further investigation to determine individual IC50 values.

Zoom Image
Fig. 2 Percent inhibition of GC376 as the positive control, THC, CBD, and CBN against SARS-CoV1 and SARS-CoV2 main protease (MPro) activities.

To examine the efficacy of THC and CBD, both candidates from our first screening study were retested. IC50 value of each cannabinoid was evaluated by equation (2) and compared to the IC50 value of GC376 ([Fig. 3 a] to [c]). CBD showed a strong effect against SARS-CoV2 main protease. In contrast, THC anticipated as an active cannabinoid in the first screening exhibited a moderate potency compared to CBD in this test.

Zoom Image
Fig. 3 Concentration-response curves of GC376 (a), CBD (b), and THC (c) against SARS-CoV2 main protease (MPro) present in normal scales.

In [Fig. 3 c], the graph demonstrated the minor activity of THC against SARS-CoV2 main protease activity with an IC50 value of 16.23 ± 1.71 µM (R2 = 0.99). In contrast, CBD exhibited intense activity against SARS-CoV2 main protease with the IC50 value of 1.86 ± 0.04 µM (R2 = 0.99). This IC50 value of CBD was only four times higher than the IC50 value of GC376 at 0.42 ± 0.06 µM (R2 = 0.98) as presented in [Fig. 3 a]. CBD test standard deviation (SD) values were more extensive than those from the THC test due to poor solubility. Higher SD values in a biological test are more frequently found for poor soluble compounds as reported by Schulz et al. (2014) and Aleo et al. (2006) [31], [32]. After combining the anti-SARS-CoV2 main protease activities and THC, CBD, and CBN chemical structures, we give a suggestion to explain the structure-activity relationship among these three (see discussion part).

Despite close structural similarity between THC, CBD, and CBN, the inhibitory activity against SARS-CoV1 and SARS-CoV2 main proteases of all three cannabinoids was significantly different. The basic core structure of the cannabinoids consists of two cyclic rings, a resorcinyl ring A and a cyclohexene ring B in all, but a dihydropyran ring C in THC and CBN only ([Fig. 4]). Molecular modelling supported that the cyclohexene substitution was most likely to have better activity than the aromatic benzene ring. Moreover, if there was a free rotation of rings A and B, the inhibitory activity seemed to increase ([Fig. 4]). This might explain why CBD showed the strongest activity among tested cannabinoids against coronavirus main proteases activity.

Zoom Image
Fig. 4 Structure-activity relationship of THC, CBD, and CBN against SARS-CoV1/2 main protease (MPro) activities compare with the actual experiment of each cannabinoid highlighting important chemical features on the cannabinoid structure. The red circle indicates an essential component on ring B, while the green circle indicates free rotation as a key feature.

After correlating the inhibitory activity of tested cannabinoids and chemical structures, we identified CBN as weakest against SARS-CoV1 and SARS-CoV2 main proteases by approximately 30% to 40%. In comparison, THC and CBD strongly inhibited both proteases completely (100%) ([Fig. 4]). Interestingly, the only difference between THC and CBN is a cyclohexene for THC and aromatic ring for CBN. We assumed that the aromatic ring B was not favorable for the inhibitory activity against SARS-CoV1 and SARS-CoV2 main proteases. We also located the essential variation between THC and CBD in the active center. Free rotation of ring B in CBD seemed to be in favor for strong activity (IC50 = 1.86 ± 0.04 µM) compared to the rigid structure in THC (IC50 = 16.23 ± 1.71 µM). This structural feature maximized the antiviral protease potency up to 8.7 times based on our experiments. We did not evaluate the IC50 value of THC and CBD against SARS-CoV1 main protease because both SARS-CoV1 and SARS-CoV2 proteases are highly identical (95%) [33].

As discussed earlier, we already knew that the aromatic ring at ring B in CBN was most likely to negatively affect the inhibitory activity ([Fig. 4], red circle). We performed the molecular docking study to understand better the possible molecular interaction between these cannabinoids and SARS-CoV2 main protease. The molecular docking hinted at the possible reason why CBN did not fit into the binding pocket of the SARS-CoV2 main protease perfectly because of the interaction between the aromatic ring and the catalytic residue, Cys145, as presented in [Fig. 5], red circle. Moreover, molecular docking supported our observation earlier in the structure-activity relationship in CBD. Docking showed that free rotation of CBD allowed the ortho-hydroxy group in ring A to establish a hydrogen bonding with His164, the amino acid in the active site, as shown in [Fig. 5], green circle. In addition, this was not present in THC. Results were in line with our in vitro experiments earlier and extended the existing knowledge regarding the molecular interaction between these cannabinoids (THC, CBD, and CBN) and SARS-CoV2 main protease.

Zoom Image
Fig. 5 Detailed 3D pose and molecular interaction of THC, CBD, CBN, and GC376 and SARS-CoV2 main protease (PDB ID: 7C6U) including the involved amino acids: a The 3D binding pose of GC376 in the binding pocket of SARS-CoV2 main protease; b Superposition 3D structures of THC, CBD, CBN, and GC376 in the binding pocket of SARS-CoV2 main protease; c The 3D binding pose of CBD; d The 3D binding pose of THC; e the 3D binding pose of CBN and SARS-CoV2 main protease; f The 2D molecular diagram of CBD; g The 2D molecular diagram of THC; h The 2D molecular diagram of CBN and the involved amino acids in the binding pocket of SARS-CoV2 main protease; A, B, and C indicates the core chemical components on the cannabinoid structure; The green and red circle indicate the key chemical feature in CBD and CBN, respectively.

Our validation showed the RMSD of 1 Å, which passed the acceptance criterion of 1.5 to 2 Å [34], [35]. With these limitations, our docking protocol provided acceptable and significant calculations to allow good predictions regarding stable confirmation and remarkable binding.

As shown in [Fig. 5 a], the binding pocket of SARS-CoV2 main protease were distinguished in six sub-pockets (S1, S2, S3, S4, S1′, and S2′). GC376 as the standard inhibitor was fitted into S1, S2, and S3 sub-pockets. The important amino acids playing a pivotal role were Cys145, the catalytic amino acid, Glu166 from the S1 sub-pocket, and Arg40, His41, Met49, Tyr54, Asp187 from the S2 sub-pocket [36]. The molecular docking results showed that THC, CBD, and CBN docked in the same position as GC376, as mentioned earlier ([Fig. 5 b] to [h]). Remarkably, CBN docking revealed that the aromatic ring interacted with Cys145, the catalytic amino acid ([Fig. 5 h], red circle). Even though the molecular docking reported that this interaction was favorable as a pi-donor hydrogen bond, we knew that this interaction must be unfavorable based on the previous in vitro experiment presented in [Fig. 5]. Having a closer look at position and poses of CBN may not fully explain less inhibitory activity ([Fig. 5 f – h]). Our docking model failed to explain reduced inhibition despite the fact that molecular interaction is quite similar to THC and CBD. Raj et al. (2021) determined stability conformation estimations of cannabinoids through density functional theory (DFT) [29]. The DFT approach was applied to determine energy gaps between HOMO and LUMO molecular orbitals. Molecules with a small frontier energy orbital gap are chemically reactive and have low kinetic stability. A higher HOMO to LUMO gap between frontier orbitals confers better stability. The authors showed that THC and CBD have slightly higher HOMO to LUMO energy gaps (ΔE 5.63, 5.80, respectively) than CBN (ΔE 4.85), which indicated that THC and CBD have a better stability profile [29], [37]. Chemical hardness (η) defined as the measure of resistance to distortion of the electron cloud represents compound stability in the binding pocket. The authors determined chemical hardness for THC and CBD (η 2.92, 2.90, respectively) being slightly higher than CBN (η 2.42) supporting again better stability for THC and CBD in the active site. Without x-ray data of co-incubated cannabinoids with the viral main protease, no final explanation can be delivered, but published data give good indications how this inhibition can be explained.

Moreover, our molecular docking supported our hypothesis why CBD had a higher activity than THC. As shown in [Fig. 5 c] and [d], the docked chemical positions between CBD and THC in the binding pocket of SARS-CoV2 main protease were different. For the CBD pose, the cyclohexene ring was pushed toward Cys145. Free rotation allowed the aromatic ring A to form a pi-donor hydrogen interaction with Gln189 from the S3 sub-pocket, as presented in a blue dash line from [Fig. 5 f]. This interaction led to a more stable hydrogen binding between the free hydroxy group in the para position of ring A and His164, the amino acid from the S1 sub-pocket presented in [Fig. 5 f], green circle. It was not confirmed for THC due to the rigid structure from ring C ([Fig. 5 g]). Moreover, this inflexible structure of THC caused an unfavorable hydrogen binding between the available hydroxyl group in ring A position and Glu166, the amino acid from the S4 sub-pocket as presented in a red dash line from [Fig. 5 g]. Finally, a summary of molecular interactions between amino acids in the active site of SARS-CoV2 main protease and compounds of interest (CBD, THC, CBN, and GC376) is provided in [Table 1], and the conserved amino acids among the compounds are given in bold.

Table 1 Molecular interactions between amino acids in the active site of the SARS-CoV2 main protease and compounds of interest (GC376, CBD, THC, and CBN).

1

2

3

4

5

6

7

8

GC376

Phe140

Gly143

Cys145

His163

His164

Glu166

CBD

Cys145

His164

Met165

Glu166

Leu167

THC

Met165

Glu166

CBN

Cys145

Met165

Consensus

3

2

3

3

9

10

11

12

13

14

15

16

GC376

Arg40

His41

Met44

Tyr54

Asp187

CBD

Pro168

His41

Met49

Gln189

THC

His41

Met49

CBN

Pro168

His41

Gln189

Consensus

2

4

2

2

After the coronaviruses attached at the hACE2 receptor, viruses are uptaken into the human host cells. Cellular replication in the human body will trigger chemokines overproduction, and proinflammatory cytokines are released we know as a cytokine storm. This storm can be downregulated by drug interaction with CB2 receptors. Eventually, this will cause organ damage, especially in the lung, as coronaviruses main target [38], [39]. Inhibition of hACE2 can slow down the viral spread in the human host or prevent viral infection [33], [40], [41]. Therefore, two active cannabinoids from earlier experiments, such as THC and CBD, were selected to evaluate the efficacy to interact with hACE2. As presented in [Fig. 6 a] to [c], we report the moderate inhibitory activity of THC and CBD against hACE2 compared to MLN-4760 as reference.

Zoom Image
Fig. 6 Concentration-response curves of MLN-4760 (a), CBD (b), and THC (c) against hACE2 enzyme present in normal scales.

The IC50 value of both THC and CBD was calculated by equation (2). The concentration-response curve ([Fig. 6]) showed the same potency for anti-hACE2 activity between THC and CBD as demonstrated by IC50 values of 11.47 ± 3.60 µM (R2 = 0.93) and 14.65 ± 0.47 µM (R2 = 0.9986) respectively. Calculated IC50 values are considered as moderate based on the previous reports [42], [43]. However, THC and CBD demonstrated a weaker activity than the reference compound MLN-4760, presented by the IC50 value of 0.03 ± 0.1 µM (R2 = 0.9899). A similar result on larger SD values was obtained from CBD again, especially at higher concentrations [44], [45]. Wang et al. (2020) reported the down-regulation effect of CBD on the hACE2 receptor on the cell surface only [28], our study has shown the effect of CBD on hACE2 as an inhibitor in combination with THC.

If hACE2 forms a complex with an inhibitor, the specific region of hACE2, namely subdomain I, will move by 13 Å from its original position, as presented in [Fig. 7 a]. This shift can be explained by the induced-fit model [46]. It is important to select the right hACE2 domain to perform the molecular docking of THC and CBD. Furthermore, the hACE2 subdomain I plays an essential role in the interaction between hACE2 and its counterpart of the viral spike protein, namely the SARS-CoV2 receptor-binding domain (SARS-CoV2 RBD) [47]. Based on this concept, it is likely that the hACE2 inhibitor can be used against SARS-CoV2 infection by interfering between hACE2 and SARS-CoV2 spike protein to prevent the virus from entering the cells [47]. Our earlier in vitro study has demonstrated moderate activity for both THC and CBD against hACE2. Therefore, molecular docking was performed to evaluate the possible binding mode of THC and CBD at the active site of hACE2. MLN-4760 as a competitive reference inhibitor was used to navigate the active site, as shown in [Fig. 7 b]. As a result, THC and CBD shared the same binding pocket with MLN-4760 at the active site of hACE2 ([Fig. 7 c]).

Zoom Image
Fig. 7 Detailed 3D pose and molecular interaction of THC, CBD, MLN-4760, and hACE2 (PDB ID: 1R4L) including the involved amino acids: a The 3D binding pose of the hACE2-MLN-4760 complex (PDB ID: 1R4L, brown color) in comparison to the hACE2-SARS-CoV2 RBD complex (PDB ID: 6LZG, pink and blue color). The narrow indicates the changing of sub-domain I between native hACE2 and hACE2-inhibitor complex, approximately 13 Å, and AS indicates the active site; b The 3D binding pose of MLN-4760 in the binding pocket of hACE2. S1 and S1′ indicates the sub-pockets in the active site; c Superposition 3D structures of THC, CBD, and MLN-4760 in the binding pocket of hACE2; d 3D binding pose of CBD; e 3D binding pose of THC; f 2D molecular diagram of CBD; g 2D molecular diagram of THC and the involved amino acids in the binding pocket of hACE2; A, B, and C indicates the core chemical components on the cannabinoid structure.

As we anticipated, our docking protocol provided an accurate outcome with a small RMSD value of about 1.2 Å, similar to the previous docking experiment on SARS-CoV2 main protease [34], [35]. Our protocol passed the acceptance criterion as mentioned earlier (see supplementary material). The same selection criteria for the selected CBD and THC poses used earlier in a previous docking experiment were also applied. The binding pocket of hACE2 can be divided into two sub-pockets S1 and S1′, as shown in [Fig. 7 b]. At least sixteen amino acids were involved in the inhibitory activity of MLN-4760, the reference, against hACE2. Five amino acids were catalytic residuals such as Arg273, His345, Pro346, Thr371, and His505. Another eight amino acids, like Glu145, Asn149, Phe274, Cys344, Met360, Cys361, Lys363, and Asp368 were from the S1′ sub-pocket. Finally, the last three amino acids from the S1 sub-pocket were Thr510, Arg514, and Try515 ([Fig. 7 b]) [46].

Our docking result suggested that THC and CBD docked at the same binding site as MLN-4760, as presented in [Fig. 7 c]. Only slight differences between CBD and THC poses were observed for ring A and its side-chain of CBD in the S1′ sub-pocket ([Fig. 7 d]). However, ring B of both cannabinoids was in similar orientation as shown in [Fig. 7 d] and [e]. Unlike the finding from SARS-CoV2 main protease, all hydroxy groups from both noncyclic (from THC) and cyclic positions (ring C of CBD) did not impact blocking hACE2 activity. The main reason is free rotation and unhindered interaction to form a hydrogen bonding with Thr371 of the catalytic residual (blue dash lines in [Fig. 7 f] and [g]). The menthenyl ring (ring B) in CBD and THC is identical, why similar molecular interactions were observed for Phe274 close to the catalytic residual of Arg273. In conclusion, there was no significant difference in terms of the molecular interaction between THC and CBD. Docking result provided supportive information suggesting why THC and CBD had the same potency in the anti-hACE2 activity. Finally, a summary of molecular interactions between amino acids in the active site of hACE2 and compounds of interest (CBD, THC, and MLN-4760 as a reference) is provided in [Table 2].

Table 2 Molecular interactions between amino acids in the active site of hACE2 and compounds of interest (MLN-4670, CBD, and THC).

1

2

3

4

5

6

7

MLN-4760

Glu145

Asn149

Arg273

Cys344

CBD

Arg273

Phe274

THC

Tyr127

Leu144

Phe274

Cys344

Consensus

2

2

2

8

9

10

11

12

13

14

MLN-4760

His345

Pro346

Cys360

Cys361

Asp368

Thr371

His374

CBD

His345

Thr371

His374

THC

His345

Pro346

Thr371

Consensus

3

2

3

2

15

16

17

18

19

20

21

MLN-4760

Glu375

His378

Glu402

His505

Tyr515

CBD

Phe504

Tyr510

THC

Consensus


#

Discussion

Based on recent reports, our data provided more insight information regarding the molecular interaction how THC and CBD inhibit the SARS-CoV2 infection in a cell-based assay [29]. In a previous study, Raj et al. (2021) reported strong antiviral activity against SARS-CoV2 infection from THC, CBD, and CBN at the same potency as remdesivir and lopinavir (clinical references) in a cell-based assay [29]. Unlike Remdesivir (a viral RNA-dependence RNA polymerase inhibitor) [48], [49] and lopinavir (a viral main protease inhibitor) [17], [50], the mechanism of THC, CBD, and CBN against SARS-CoV2 infection are still not fully understood. Only the predicted mechanism as an inhibitor against viral protease was proposed from computational experiments [29], [30]. Our experiments showed that THC and CBD might inhibit SARS-CoV1 and SARS-CoV2 main proteases but not CBN. Therefore, we partially agree with the predicted mechanism from previous studies and propose the hypothesis that CBN may have a different mechanism of action against SARS-CoV2 infection. Notably, we evaluated the structure-activity relationship among THC, CBD, and CBN against SARS-CoV2 main protease. Two major chemical features that are likely to play an essential role in this inhibitory concept are the cyclohexene ring B and the free rotation between ring A and B. Remarkably, THC and CBD are not just only inhibiting the viral proteases like lopinavir, but also impact hACE2 interaction.

We have not discussed any activity of THC, CBD and CBN as potent CB2 agonists know as important target to reduce the proinflammatory effect of SARS-CoV2. THC is known as a partial agonist, as well as CBN being agonist and inverse agonist. If we also consider the pharmacological level of CB receptor-mediated effects of the endocannabinoid system, we see that cannabinoids can probably take a major role in the therapy of SARS-COv2 infections. It is exciting to see that a systems pharmacological approach gives us the confidence that we donʼt have to deal with just THC, CBD and CBN. We know that more than 130 cannabinoids are known in the cannabis plant and the function and pharmacological action of the minor cannabinoids like CBC, CBL, CBE gives needs to be explored. We hope that prenylated terpenophenolics being the typical scaffold of cannabinoids become an interesting class of future antiviral agents.


#

Material and Methods

Chemicals

THC and CBD (both purity ≥ 98%) were purchased from Cayman Chemical Company, but CBN was synthesized in our laboratory. The synthesis routine of CBN (purity ≥ 98%) is explained below. The work is carried out with permission of the Federal German Opium Agency (4 586 416).

Reaction: to a solution of CBD (314 mg, 1.0 mmol, 1.0 eq) in toluene (30 mL) iodine was added (634.5 mg, 2.5 mmol, 2.5 eq) under nitrogen. The mixture was refluxed for 2 h. After then, the reaction was quenched by cooling to room temperature and sequentially washed with 20 mL of sodium thiosulfate 10% (Na2S2O3). The organic layers were washed by brine and water. After drying through magnesium sulfate, the organic phase was evaporated, and the residue was purified with flash column chromatography (ethyl acetate: petroleum ether = 1 : 99 to 3 : 97) to afford CBN as a pale red oil (189.1 mg, 61%).

TLC: EtOAc : petroleum ether = 9 : 1; Rf-CBD = 0.45, Rf-CBN = 0.35.

The nuclear magnetic resonance or NMR: 1H NMR (400 MHz; CDCl3): δ = 0.89 (t, 3H, J = 7.2 Hz), 1.28 – 1.33 (m, 4H), 1.60 (s, 6H), 1.61 – 1.64 (m, 2H), 2.31 (s, 3H) 2.40 (t, 2H, J = 7.3 Hz), 5.30 (br, 1H, -OH), 6.26 (s,1H), 6.41 (s, 1H,), 7.02 (d, 1H, J = 7.4 Hz), 7.11 (d, 1H, J = 7.8 Hz), 8.18 (s, 1H). 13C NMR (100 MHz; CDCl3): δ = 14.3, 21.4, 22.7, 27.3, 30.6, 31.6, 35.8, 87.4, 108.6,110.0, 110.2, 111.1, 122.4, 126.2, 127.3, 127.6, 137.3, 144.2, 153.3, 154.9. 1H/13C-NMR spectrogram of CBN to be found in the supplementary file.


#

Inhibitory activity of SARS-CoV1 and SARS-CoV2 Main Proteases

The assays were performed by using the SARS-CoV1 and SARS-CoV2 protease assay kits from BPS Bioscience. The authors performed these assays by following the manufactory guideline strictly. The used protocols were summarized here. Firstly, the proper concentration of SARS-CoV1 and SARS-CoV2 protease was adjusted to 5 µg/mL and 4 µg/mL, respectively as recommended by adding 1 mM DTT assay buffer. Secondly, 30 µl of the protease solution was mixed with 10 µl of 0.5 mg/mL sample solution or 500 µM GC376 solution for the positive control or 5% DMSO for the negative control. Thirdly, these mixtures were incubated at room temperature for 30 minutes before adding 10 µl of 10 µM fluorogenic substrate solution. Finally, the reaction was read by a FLUOStar Omega microplate reader at λexcitation/emission of 360 nm/460 nm after 4 hours and overnight incubation times for SARS-CoV1 and SARS-CoV2 protease assays consequently. Each cannabinoid was studied in triplicate. The percent inhibition was calculated after the background color elimination. The equation that was used to calculate the percent inhibition of each sample was presented below as equation (1).

% Inhibition = ((FIcontrol – FItest sample)/FIcontrol) × 100(1)

When FI stands for a fluorescence intensity that was read by the microplate reader and the percent inhibition of each sample was presented in a standard form of mean ± SD values.

All cannabinoids that showed a percent inhibition higher than 80% were subjected to determine a half inhibitory activity concentration or IC50 to evaluate their potencies. Five concentrations of each active cannabinoid in a range of 318 µM to 4 µM were prepared by half fold. Finally, the IC50 value was calculated by the second equation or equation (2), which was described below.

% Inhibition = 100/(1+(IC50/Concentration)Hill)(2)

When concentration referred to the concentration providing the percent inhibition and Hill value referred to the Hill coefficient and the obtained IC50 values of the active cannabinoids were compared to the IC50 value of GC376, the standard control provided by BPS Bioscience.


#

Inhibitory activity against hACE2

The anti-hACE2 activity was performed only with the active cannabinoids from the earlier experiment using the ACE2 inhibitor screening assay kit from BPS Bioscience. The assay protocol from the manufacturing was applied here strictly and would be explained shortly. In the beginning, the concentration of hACE2 was diluted by the assay buffer until reaching the desire concentration of 0.5 µg/mL. After that, 20 µl of the prepared hACE2 solution as mention earlier was mixed with 5 µl of 10 mg/mL sample solution or 10% DMSO solution as a control. Then, incubation of 15 minutes was applied at room temperature. Finally, 25 µl of hACE2 fluorogenic substrate solution was added to the wells. The reaction was read after 1 hour incubation time by the same microplate reader as mentioned in the previous section λexcitation/emission of 555 nm/585 nm. Each sample was tested three times. The IC50 value of the cannabinoids was also calculated by equation 2 as presented earlier. Once again, the IC50 value of MLN-4760 as a positive control was provided by BPS Bioscience.


#

Statistical analysis

The descriptive statistics from Microsoft Excel 365 such as mean, and SD were used to calculate the percent inhibition from equation (1) and the R2 value from equation (2) was also calculated by the correlation function from MS Excel. Moreover, all graphs in this study were generated by Microsoft Excel as well [51], [52]. First, the solver Add-in package from Microsoft Excel was applied to calculate an initial IC50 value from equation (2) [53], [54]. Finally, the nonlinear least squares (nls) function from RStudio (version 1.4.1717) was used to fit the model and determine all parameters (IC50, standard error (SE), 95% confident interval (95%CI), Hill coefficient, and R2) [55], [56], [57], [58].


#

In silico study by molecular docking

3D cannabinoids chemical structures preparation

All cannabinoid chemical structures were downloaded from the PubChem database (https://pubchem-ncbi-nlm-nih-gov.accesdistant.sorbonne-universite.fr/), such as THC (PubChem CID: 16 078), CBD (PubChem CID: 644 019), and CBN (PubChem CID: 2543). The geometric optimization and MMFF94 energetic minimization of all cannabinoids were performed by Avogadro version 1.2.0 [59]. All optimized cannabinoids were subjected to AutoDockTools version 1.5.6 for the final preparation before performing the molecular docking experiment [60].


#

Preparation of SARS-CoV2 main protease, SARS-CoV2 receptor-binding domain (SARS-CoV2 RBD), hACE2 proteins and their native ligands

All of the crystal structures as mention above were obtained from the RSCB protein databank (https://www.rcsb.org/) such as SARS-CoV2 main protease with GC376 (PDB ID: 7C6U) [36], SARS-CoV2 RBD in complex with hACE2 (PDB ID: 6LZG) [47], and hACE2 with MLN-4760 (PDB ID: 1R4L) [46]. USFC Chimera version 1.11.2 was used to remove water molecules from the crystal structures and extract the native ligand. The inhibitor came with the crystal structure was used to validate the established docking protocol later [61]. Once again, Autodock Tools from the earlier section was applied here to prepare these protein structures and their extracted native ligand before the docking experiment [60].


#

Molecular docking experiment

AutoDock Vina version 1.1.2 was selected to perform the docking experiment and the binding pocket (active site) of each protein was guided by the native ligand as mention above. Each protein had a specific coordinate for its binding pocket as also known as grid box, which was demonstrated in terms of the x, y, and z coordinate system [60]. The grid box of SARS-CoV2 main protease (PDB ID: 7C6U) was set as x = − 21.1, y = − 26.5 and z = 2.3, while the grid box of hACE2 was set as x = 40.2, y = 6.0 and z = 29.0. Even though these coordinates were different, but the size of these grid boxes was set the same as 18 Å × 18 Å × 18 Å. The other docking parameters were set as default [60]. Before applying these established docking protocols to examine the binding mode of the cannabinoids as described above, these protocols had to be validated by redocking the extracted native ligand back into its original position in the protein. Only the protocol that imitated the original pose of the native ligand with root-mean-square deviation (RMSD) less than 2 Å compared before and after redocking was used in this study [34], [62], [63], [64]. The docking pose was selected using the combination of four main criteria and defined as the best pose. First, native ligands such as GC376 and MLN-4760 were used as a reference. Second, docking poses with no hydrogen bonding were excluded. Third, only a pose that exhibited the lowest binding energy was selected. Finally, the docking pose of each cannabinoid was selected independently [65], [66].


#

Post-docking analysis

The ViewDock package visualized the docking results of each cannabinoid from the Chimera program as mentioned earlier [61]. For the interaction diagram, Discovery Studio Visualizer free version was applied [67].


#
#
#

Contributorsʼ Statement

Conceptualization: T. Pitakbut and O. Kayser; investigation both in vitro and docking experiments: T. Pitakbut; chemical synthesis and structure elucidation: GN. Nguyen; writing, original draft preparation: T. Pitakbut; writing, review and editing: O. Kayser; supervision: O. Kayser. All authors have read and agreed to the published version of the manuscript.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgements

We acknowledge financial support by the German Academic Exchange Service (DAAD), grant number 57 299 294.

Supporting Information

  • References

  • 1 Ben-Shabat S, Yarmolinsky L, Porat D, Dahan A. Antiviral effect of phytochemicals from medicinal plants: Applications and drug delivery strategies. Drug Deliv Transl Res 2020; 10: 354-367
  • 2 Watanabe K. Drug-repositioning approach for the discovery of anti-influenza virus activity of Japanese herbal (Kampo) medicines in vitro: Potent high activity of Daio-Kanzo-To. Evid Based Complement Alternat Med 2018; 2018: 6058181
  • 3 Rajasekaran D, Palombo EA, Chia Yeo T, Lim Siok Ley D, Lee Tu C, Malherbe F, Grollo L. Identification of traditional medicinal plant extracts with novel anti-influenza activity. PLoS One 2013; 8: e79293
  • 4 Parvez MK, Tabish Rehman M, Alam P, Al-Dosari MS, Alqasoumi SI, Alajmi MF. Plant-derived antiviral drugs as novel hepatitis B virus inhibitors: Cell culture and molecular docking study. Saudi Pharm J 2019; 27: 389-400
  • 5 Jahan I, Onay A. Potentials of plant-based substance to inhabit and probable cure for the COVID-19. Turk J Biol 2020; 44: 228-241
  • 6 Boukhatem MN, Setzer WN. Aromatic herbs, medicinal plant-derived essential oils, and phytochemical extracts as potential therapies for Coronaviruses: Future perspectives. Plants 2020; 9: 800
  • 7 Wink M. Potential of DNA intercalating alkaloids and other plant secondary metabolites against SARS-CoV-2 causing COVID-19. Diversity 2020; 12: 175
  • 8 Martinez JP, Sasse F, Brönstrup M, Diez J, Meyerhans A. Antiviral drug discovery: Broad-spectrum drugs from nature. Nat Prod Rep 2015; 32: 29-48
  • 9 Hensel A, Bauer R, Heinrich M, Spiegler V, Kayser O, Hempel G, Kraft K. Challenges at the time of COVID-19: Opportunities and innovations in antivirals from nature. Planta Med 2020; 86: 659-664
  • 10 De Clercq E, Li G. Approved antiviral drugs over the past 50 years. Clin Microbiol Rev 2016; 29: 695-747
  • 11 Wang Z, Yang L. Turning the tide: Natural products and natural-product-inspired chemicals as potential counters to SARS-CoV-2 infection. Front Pharmacol 2020; 11: 1013
  • 12 Ni D, Ho DH, Vijjeswarapu M, Felix E, Rhea PR, Newman RA. Metabolism of homoharringtonine, a cytotoxic component of the evergreen plant Cephalotaxus harringtonia. J Exp Ther Oncol 2003; 3: 47-52
  • 13 Abdelkafi H, Nay B. Natural products from Cephalotaxus sp.: chemical diversity and synthetic aspects. Nat Prod Rep 2012; 29: 845-869
  • 14 Kaur P, Thiruchelvan M, Lee RCH, Chen H, Chen KC, Ng ML, Chu JJ. Inhibition of chikungunya virus replication by harringtonine, a novel antiviral that suppresses viral protein expression. Antimicrob Agents Chemother 2013; 57: 155
  • 15 Dong HJ, Wang ZH, Meng W, Li CC, Hu YX, Zhou L, Wang XJ. The natural compound homoharringtonine presents broad antiviral activity in vitro and in vivo . Viruses 2018; 10: 601
  • 16 Kim JE, Song YJ. Anti-varicella-zoster virus activity of cephalotaxine esters in vitro . J Microbiol 2019; 57: 74-79
  • 17 Choy KT, Wong AYL, Kaewpreedee P, Sia SF, Chen D, Hui KPY, Chu DKW, Chan MCW, Cheung PP, Huang X, Peiris M, Yen HL. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro . Antiviral Res 2020; 178: 104786
  • 18 Bleasel MD, Peterson GM. Emetine, Ipecac, Ipecac alkaloids and analogues as potential antiviral agents for Coronaviruses. Pharmaceuticals 2020; 13: 51
  • 19 Rosales-López C, Muñoz-Arrieta R, Abdelnour-Esquivel A. Emetine and cephaeline content in plants of Psychotria ipecacuanha in Costa Rica. Rev Colomb Quim 2020; 49: 18-22
  • 20 Yang S, Xu M, Lee EM, Gorshkov K, Shiryaev SA, He S, Sun W, Cheng YS, Hu X, Tharappel AM, Lu B, Pinto A, Farhy C, Huang CT, Zhang Z, Zhu W, Wu Y, Zhou Y, Song G, Zhu H, Shamim K, Martínez-Romero C, García-Sastre A, Preston RA, Jayaweera DT, Huang R, Huang W, Xia M, Simeonov A, Ming G, Qiu X, Terskikh AV, Tang H, Song H, Zheng W. Emetine inhibits Zika and Ebola virus infections through two molecular mechanisms: Inhibiting viral replication and decreasing viral entry. Cell Discov 2018; 4: 31
  • 21 Wang A, Sun Y, Liu Q, Wu H, Liu J, He J, Yu J, Chen QQ, Ge Y, Zhang Z, Hu C, Chen C, Qi Z, Zou F, Liu F, Hu J, Zhao M, Huang T, Wang B, Wang L, Wang W, Wang W, Ren T, Liu J, Sun Y, Fan S, Wu Q, Liang C, Sun L, Su B, Wei W, Liu Q. Low dose of emetine as potential anti-SARS-CoV-2 virus therapy: Preclinical in vitro inhibition and in vivo pharmacokinetic evidences. Mol biomed 2020; 1: 14
  • 22 Ribaudo G, Coghi P, Yang LJ, Ng JPL, Mastinu A, Memo M, Wong VKW, Gianoncelli A. Computational and experimental insights on the interaction of artemisinin, dihydroartemisinin and chloroquine with SARS-CoV-2 spike protein receptor-binding domain (RBD). Nat Prod Res 2021;
  • 23 El Biali M, Broers B, Besson M, Demeules J. Cannabinoids and COVID-19. Med Cannabis Cannabinoids 2020; 3: 111-115
  • 24 Hill KP. Cannabinoids and the Coronavirus. Cannabis Cannabinoid Res 2020; 5: 118-120
  • 25 Esposito G, Pesce M, Seguella L, Sanseverino W, Lu J, Corpetti C, Sarnelli G. The potential of cannabidiol in the COVID-19 pandemic. Br J Pharmacol 2020; 177: 4967-4970
  • 26 Onaivi ES, Sharma V. Cannabis for COVID-19: can cannabinoids quell the cytokine storm?. Future Sci OA 2020; 6: FSO625
  • 27 Dzobo K, Chiririwa H, Dandara C, Dzobo W. Coronavirus disease-2019 treatment strategies targeting interleukin-6 signaling and herbal medicine. OMICS 2020; 25: 13-22
  • 28 Wang B, Kovalchuk A, Li D, Rodriguez-Juarez R, Ilnytskyy Y, Kovalchuk I, Kovalchuk O. In search of preventive strategies: Novel high-CBD Cannabis sativa extracts modulate ACE2 expression in COVID-19 gateway tissues. Aging (Albany NY) 2020; 12: 22425-22444
  • 29 Raj V, Park JG, Cho KH, Choi P, Kim T, Ham J, Lee J. Assessment of antiviral potencies of cannabinoids against SARS-CoV-2 using computational and in vitro approaches. Int J Biol Macromol 2021; 168: 474-485
  • 30 Tallei TE, Tumilaar SG, Niode NJ, Fatimawali. Kepel BJ, Idroes R, Effendi Y, Sakib SA, Emran TB. Potential of plant bioactive compounds as SARS-CoV-2 Main Protease (Mpro) and Spike (S) glycoprotein inhibitors: A molecular docking study. Scientifica (Cairo) 2020; 2020: 6307457
  • 31 Schulz U, Freitag M, Schmidt K, Witetschek M, Polzin M, Morgenstern O. Synthesis and structure elucidation of 2,3,5,6,7,8-hexahydro-1 H-[1,2,4]triazolo[1,2-a]pyridazine-1-thione, 3,3-disubstituted and 2-substituted derivatives and evaluation of their inhibitory activity against inducible nitric oxide synthase. Pharmazie 2014; 69: 731-744
  • 32 Aleo MF, Bettoni F, Boniotti J, Morandini F, Giuliani R, Steimberg N, Apostoli P, Mazzoleni G. A comparative in vitro study of the toxic potency of five inorganic lead compounds on a rat liver epithelial cell line (REL). Toxicol In Vitro 2006; 20: 874-881
  • 33 Chitranshi N, Gupta VK, Rajput R, Godinez A, Pushpitha K, Shen T, Mirzaei M, You Y, Basavarajappa D, Gupta V, Graham SL. Evolving geographic diversity in SARS-CoV2 and in silico analysis of replicating enzyme 3 CLpro targeting repurposed drug candidates. J Transl Med 2020; 18: 278
  • 34 Hevener KE, Zhao W, Ball DM, Babaoglu K, Qi J, White SW, Lee RE. Validation of molecular docking programs for virtual screening against dihydropteroate synthase. J Chem Inf Model 2009; 49: 444-460
  • 35 Pagadala NS, Syed K, Tuszynski J. Software for molecular docking: A review. Biophys Rev 2017; 9: 91-102
  • 36 Fu L, Ye F, Feng Y, Yu F, Wang Q, Wu Y, Zhao C, Sun H, Huang B, Niu P, Song H, Shi Y, Li X, Tan W, Qi J, Gao GF. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat Commun 2020; 11: 4417
  • 37 Böhme T, Simpson CD, Müllen K, Rabe JP. Current–voltage characteristics of a homologous series of polycyclic aromatic hydrocarbons. Chem Eur J 2007; 13: 7349-7357
  • 38 Tay MZ, Poh CM, Rénia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol 2020; 20: 363-374
  • 39 Rossi F, Tortora C, Argenziano M, Di Paola A, Punzo F. Cannabinoid receptor type 2: A possible target in SARS-CoV-2 (CoV-19) infection?. Int J Mol Sci 2020; 21: 3809
  • 40 Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and Is blocked by a clinically proven protease inhibitor. Cell 2020; 181: 271-280.e8
  • 41 Jin Z, Du X, Xu Y, Deng Y, Liu M, Zhao Y, Zhang B, Li X, Zhang L, Peng C, Duan Y, Yu J, Wang L, Yang K, Liu F, Jiang R, Yang X, You T, Liu X, Yang X, Bai F, Liu H, Liu X, Guddat LW, Xu W, Xiao G, Qin C, Shi Z, Jiang H, Rao Z, Yang H. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020; 582: 289-293
  • 42 Choi BW, Lee HS, Shin HC, Lee BH. Multifunctional activity of polyphenolic compounds associated with a potential for alzheimerʼs disease therapy from Ecklonia cava. Phytother Res 2015; 29: 549-553
  • 43 Rana M, Pareek A, Bhardwaj S, Arya G, Nimesh S, Arya H, Bhatt TK, Yaragorla S, Sharma AK. Aryldiazoquinoline based multifunctional small molecules for modulating Aβ42 aggregation and cholinesterase activity related to Alzheimerʼs disease. RSC Adv 2020; 10: 28827-28837
  • 44 Koch N, Jennotte O, Gasparrini Y, Vandenbroucke F, Lechanteur A, Evrard B. Cannabidiol aqueous solubility enhancement: Comparison of three amorphous formulations strategies using different type of polymers. Int J Pharm 2020; 589: 119812
  • 45 Millar SA, Maguire RF, Yates AS, OʼSullivan SE. Towards better delivery of Cannabidiol (CBD). Pharmaceuticals (Basel) 2020; 13: 219
  • 46 Towler P, Staker B, Prasad SG, Menon S, Tang J, Parsons T, Ryan D, Fisher M, Williams D, Dales NA, Patane MA, Pantoliano MW. ACE2 X-ray structures reveal a large hinge-bending motion important for inhibitor binding and catalysis. J Biol Chem 2004; 279: 17996-18007
  • 47 Wang Q, Zhang Y, Wu L, Niu S, Song C, Zhang Z, Lu G, Qiao C, Hu Y, Yuen KY, Wang Q, Zhou H, Yan J, Qi J. Structural and functional basis of SARS-CoV-2 entry by using human ACE2. Cell 2020; 181: 894-904.e9
  • 48 Malin JJ, Suárez I, Priesner V, Fätkenheuer G, Rybniker J. Remdesivir against COVID-19 and other viral diseases. Clin Microbiol Rev 2020; 34: e00162-20
  • 49 Gordon CJ, Tchesnokov EP, Woolner E, Perry JK, Feng JY, Porter DP, Götte M. Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency. J Biol Chem 2020; 295: 6785-6797
  • 50 Maciorowski D, Idrissi SZE, Gupta Y, Medernach BJ, Burns MB, Becker DP, Durvasula R, Kempaiah P. A review of the preclinical and clinical efficacy of remdesivir, hydroxychloroquine, and lopinavir-ritonavir treatments against COVID-19. SLAS Discov 2020; 25: 1108-1122
  • 51 Carr NT. Using Microsoft Excel® to calculate descriptive statistics and create graphs. Lang Assess Q 2008; 5: 43-62
  • 52 Papautsky EL, Hamlish T. Patient-reported treatment delays in breast cancer care during the COVID-19 pandemic. Breast Cancer Res Treat 2020; 184: 249-254
  • 53 Becker RC. COVID-19 and its sequelae: a platform for optimal patient care, discovery and training. J Thromb Thrombolysis 2021;
  • 54 Jasper EE, Ajibola VO, Onwuka JC. Nonlinear regression analysis of the sorption of crystal violet and methylene blue from aqueous solutions onto an agro-waste derived activated carbon. Appl Water Sci 2020; 10: 132
  • 55 R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2021. Accessed March 16 2021 at: https://www.R-project.org/
  • 56 R Studio Team. RStudio: Integrated Development Environment for R. Boston, MA: RStudio, PBC; 2021. Accessed March 16, 2021 at: http://www.rstudio.com/
  • 57 Baty F, Ritz C, Charles S, Brutsche M, Flandrois JP, Delignette-Muller ML. A toolbox for nonlinear regression in R: The package nlstools. J Stat Softw 2015; 66: 1-21
  • 58 Zeviani WM. wzRfun: Walmes Zevianiʼs collection of functions. 2019. Accessed March 18, 2021 at: https://github.com/walmes/wzRfun
  • 59 Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, Hutchison GR. Avogadro: An advanced semantic chemical editor, visualization, and analysis platform. J Cheminformatics 2012; 4: 17
  • 60 Trott O, Olson AJ. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31: 455-461
  • 61 Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE. UCSF Chimera–A visualization system for exploratory research and analysis. J Comput Chem 2004; 25: 1605-1612
  • 62 Schneider N, Hindle S, Lange G, Klein R, Albrecht J, Briem H, Beyer K, Claußen H, Gastreich M, Lemmen C, Rarey M. Substantial improvements in large-scale redocking and screening using the novel HYDE scoring function. J Comput Aided Mol Des 2012; 26: 701-723
  • 63 Musoev A, Numonov S, You Z, Gao H. Discovery of novel DPP-IV inhibitors as potential candidates for the treatment of type 2 diabetes mellitus predicted by 3D QSAR Pharmacophore models, molecular docking and de novo evolution. Molecules 2019; 24: 2870
  • 64 Gurung AB, Ali MA, Lee J, Farah MA, Al-Anazi KM. Unravelling lead antiviral phytochemicals for the inhibition of SARS-CoV-2 Mpro enzyme through in silico approach. Life Sci 2020; 255: 117831
  • 65 Ramírez D, Caballero J. Is it reliable to take the molecular docking top scoring position as the best solution without considering available structural data?. Molecules 2018; 23: 1038
  • 66 Velázquez-Libera JL, Murillo-López JA, de la Torre AF, Caballero J. Structural requirements of N-alpha-Mercaptoacetyl Dipeptide (NAMdP) inhibitors of pseudomonas aeruginosa virulence factor LasB: 3D-QSAR, molecular docking, and interaction fingerprint studies. Int J Mol Sci 2019; 20: 6133
  • 67 Biovia DS. Discovery Studio Visualizer. San Diego, CA, USA: Dassault Systèmes; 2017: 936

Correspondence

Prof. Dr. Dr. h. c. Oliver Kayser
Technical Biochemistry
Faculty of Biochemical and Chemical Engineering
TU Dortmund University
Emil-Figge-Strasse 66
44227 Dortmund
Germany   
Phone: + 49 23 17 55 74 87   
Fax: + 49 23 17 55 74 89   

Publication History

Received: 01 May 2021

Accepted after revision: 03 August 2021

Article published online:
12 October 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Ben-Shabat S, Yarmolinsky L, Porat D, Dahan A. Antiviral effect of phytochemicals from medicinal plants: Applications and drug delivery strategies. Drug Deliv Transl Res 2020; 10: 354-367
  • 2 Watanabe K. Drug-repositioning approach for the discovery of anti-influenza virus activity of Japanese herbal (Kampo) medicines in vitro: Potent high activity of Daio-Kanzo-To. Evid Based Complement Alternat Med 2018; 2018: 6058181
  • 3 Rajasekaran D, Palombo EA, Chia Yeo T, Lim Siok Ley D, Lee Tu C, Malherbe F, Grollo L. Identification of traditional medicinal plant extracts with novel anti-influenza activity. PLoS One 2013; 8: e79293
  • 4 Parvez MK, Tabish Rehman M, Alam P, Al-Dosari MS, Alqasoumi SI, Alajmi MF. Plant-derived antiviral drugs as novel hepatitis B virus inhibitors: Cell culture and molecular docking study. Saudi Pharm J 2019; 27: 389-400
  • 5 Jahan I, Onay A. Potentials of plant-based substance to inhabit and probable cure for the COVID-19. Turk J Biol 2020; 44: 228-241
  • 6 Boukhatem MN, Setzer WN. Aromatic herbs, medicinal plant-derived essential oils, and phytochemical extracts as potential therapies for Coronaviruses: Future perspectives. Plants 2020; 9: 800
  • 7 Wink M. Potential of DNA intercalating alkaloids and other plant secondary metabolites against SARS-CoV-2 causing COVID-19. Diversity 2020; 12: 175
  • 8 Martinez JP, Sasse F, Brönstrup M, Diez J, Meyerhans A. Antiviral drug discovery: Broad-spectrum drugs from nature. Nat Prod Rep 2015; 32: 29-48
  • 9 Hensel A, Bauer R, Heinrich M, Spiegler V, Kayser O, Hempel G, Kraft K. Challenges at the time of COVID-19: Opportunities and innovations in antivirals from nature. Planta Med 2020; 86: 659-664
  • 10 De Clercq E, Li G. Approved antiviral drugs over the past 50 years. Clin Microbiol Rev 2016; 29: 695-747
  • 11 Wang Z, Yang L. Turning the tide: Natural products and natural-product-inspired chemicals as potential counters to SARS-CoV-2 infection. Front Pharmacol 2020; 11: 1013
  • 12 Ni D, Ho DH, Vijjeswarapu M, Felix E, Rhea PR, Newman RA. Metabolism of homoharringtonine, a cytotoxic component of the evergreen plant Cephalotaxus harringtonia. J Exp Ther Oncol 2003; 3: 47-52
  • 13 Abdelkafi H, Nay B. Natural products from Cephalotaxus sp.: chemical diversity and synthetic aspects. Nat Prod Rep 2012; 29: 845-869
  • 14 Kaur P, Thiruchelvan M, Lee RCH, Chen H, Chen KC, Ng ML, Chu JJ. Inhibition of chikungunya virus replication by harringtonine, a novel antiviral that suppresses viral protein expression. Antimicrob Agents Chemother 2013; 57: 155
  • 15 Dong HJ, Wang ZH, Meng W, Li CC, Hu YX, Zhou L, Wang XJ. The natural compound homoharringtonine presents broad antiviral activity in vitro and in vivo . Viruses 2018; 10: 601
  • 16 Kim JE, Song YJ. Anti-varicella-zoster virus activity of cephalotaxine esters in vitro . J Microbiol 2019; 57: 74-79
  • 17 Choy KT, Wong AYL, Kaewpreedee P, Sia SF, Chen D, Hui KPY, Chu DKW, Chan MCW, Cheung PP, Huang X, Peiris M, Yen HL. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro . Antiviral Res 2020; 178: 104786
  • 18 Bleasel MD, Peterson GM. Emetine, Ipecac, Ipecac alkaloids and analogues as potential antiviral agents for Coronaviruses. Pharmaceuticals 2020; 13: 51
  • 19 Rosales-López C, Muñoz-Arrieta R, Abdelnour-Esquivel A. Emetine and cephaeline content in plants of Psychotria ipecacuanha in Costa Rica. Rev Colomb Quim 2020; 49: 18-22
  • 20 Yang S, Xu M, Lee EM, Gorshkov K, Shiryaev SA, He S, Sun W, Cheng YS, Hu X, Tharappel AM, Lu B, Pinto A, Farhy C, Huang CT, Zhang Z, Zhu W, Wu Y, Zhou Y, Song G, Zhu H, Shamim K, Martínez-Romero C, García-Sastre A, Preston RA, Jayaweera DT, Huang R, Huang W, Xia M, Simeonov A, Ming G, Qiu X, Terskikh AV, Tang H, Song H, Zheng W. Emetine inhibits Zika and Ebola virus infections through two molecular mechanisms: Inhibiting viral replication and decreasing viral entry. Cell Discov 2018; 4: 31
  • 21 Wang A, Sun Y, Liu Q, Wu H, Liu J, He J, Yu J, Chen QQ, Ge Y, Zhang Z, Hu C, Chen C, Qi Z, Zou F, Liu F, Hu J, Zhao M, Huang T, Wang B, Wang L, Wang W, Wang W, Ren T, Liu J, Sun Y, Fan S, Wu Q, Liang C, Sun L, Su B, Wei W, Liu Q. Low dose of emetine as potential anti-SARS-CoV-2 virus therapy: Preclinical in vitro inhibition and in vivo pharmacokinetic evidences. Mol biomed 2020; 1: 14
  • 22 Ribaudo G, Coghi P, Yang LJ, Ng JPL, Mastinu A, Memo M, Wong VKW, Gianoncelli A. Computational and experimental insights on the interaction of artemisinin, dihydroartemisinin and chloroquine with SARS-CoV-2 spike protein receptor-binding domain (RBD). Nat Prod Res 2021;
  • 23 El Biali M, Broers B, Besson M, Demeules J. Cannabinoids and COVID-19. Med Cannabis Cannabinoids 2020; 3: 111-115
  • 24 Hill KP. Cannabinoids and the Coronavirus. Cannabis Cannabinoid Res 2020; 5: 118-120
  • 25 Esposito G, Pesce M, Seguella L, Sanseverino W, Lu J, Corpetti C, Sarnelli G. The potential of cannabidiol in the COVID-19 pandemic. Br J Pharmacol 2020; 177: 4967-4970
  • 26 Onaivi ES, Sharma V. Cannabis for COVID-19: can cannabinoids quell the cytokine storm?. Future Sci OA 2020; 6: FSO625
  • 27 Dzobo K, Chiririwa H, Dandara C, Dzobo W. Coronavirus disease-2019 treatment strategies targeting interleukin-6 signaling and herbal medicine. OMICS 2020; 25: 13-22
  • 28 Wang B, Kovalchuk A, Li D, Rodriguez-Juarez R, Ilnytskyy Y, Kovalchuk I, Kovalchuk O. In search of preventive strategies: Novel high-CBD Cannabis sativa extracts modulate ACE2 expression in COVID-19 gateway tissues. Aging (Albany NY) 2020; 12: 22425-22444
  • 29 Raj V, Park JG, Cho KH, Choi P, Kim T, Ham J, Lee J. Assessment of antiviral potencies of cannabinoids against SARS-CoV-2 using computational and in vitro approaches. Int J Biol Macromol 2021; 168: 474-485
  • 30 Tallei TE, Tumilaar SG, Niode NJ, Fatimawali. Kepel BJ, Idroes R, Effendi Y, Sakib SA, Emran TB. Potential of plant bioactive compounds as SARS-CoV-2 Main Protease (Mpro) and Spike (S) glycoprotein inhibitors: A molecular docking study. Scientifica (Cairo) 2020; 2020: 6307457
  • 31 Schulz U, Freitag M, Schmidt K, Witetschek M, Polzin M, Morgenstern O. Synthesis and structure elucidation of 2,3,5,6,7,8-hexahydro-1 H-[1,2,4]triazolo[1,2-a]pyridazine-1-thione, 3,3-disubstituted and 2-substituted derivatives and evaluation of their inhibitory activity against inducible nitric oxide synthase. Pharmazie 2014; 69: 731-744
  • 32 Aleo MF, Bettoni F, Boniotti J, Morandini F, Giuliani R, Steimberg N, Apostoli P, Mazzoleni G. A comparative in vitro study of the toxic potency of five inorganic lead compounds on a rat liver epithelial cell line (REL). Toxicol In Vitro 2006; 20: 874-881
  • 33 Chitranshi N, Gupta VK, Rajput R, Godinez A, Pushpitha K, Shen T, Mirzaei M, You Y, Basavarajappa D, Gupta V, Graham SL. Evolving geographic diversity in SARS-CoV2 and in silico analysis of replicating enzyme 3 CLpro targeting repurposed drug candidates. J Transl Med 2020; 18: 278
  • 34 Hevener KE, Zhao W, Ball DM, Babaoglu K, Qi J, White SW, Lee RE. Validation of molecular docking programs for virtual screening against dihydropteroate synthase. J Chem Inf Model 2009; 49: 444-460
  • 35 Pagadala NS, Syed K, Tuszynski J. Software for molecular docking: A review. Biophys Rev 2017; 9: 91-102
  • 36 Fu L, Ye F, Feng Y, Yu F, Wang Q, Wu Y, Zhao C, Sun H, Huang B, Niu P, Song H, Shi Y, Li X, Tan W, Qi J, Gao GF. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat Commun 2020; 11: 4417
  • 37 Böhme T, Simpson CD, Müllen K, Rabe JP. Current–voltage characteristics of a homologous series of polycyclic aromatic hydrocarbons. Chem Eur J 2007; 13: 7349-7357
  • 38 Tay MZ, Poh CM, Rénia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol 2020; 20: 363-374
  • 39 Rossi F, Tortora C, Argenziano M, Di Paola A, Punzo F. Cannabinoid receptor type 2: A possible target in SARS-CoV-2 (CoV-19) infection?. Int J Mol Sci 2020; 21: 3809
  • 40 Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and Is blocked by a clinically proven protease inhibitor. Cell 2020; 181: 271-280.e8
  • 41 Jin Z, Du X, Xu Y, Deng Y, Liu M, Zhao Y, Zhang B, Li X, Zhang L, Peng C, Duan Y, Yu J, Wang L, Yang K, Liu F, Jiang R, Yang X, You T, Liu X, Yang X, Bai F, Liu H, Liu X, Guddat LW, Xu W, Xiao G, Qin C, Shi Z, Jiang H, Rao Z, Yang H. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020; 582: 289-293
  • 42 Choi BW, Lee HS, Shin HC, Lee BH. Multifunctional activity of polyphenolic compounds associated with a potential for alzheimerʼs disease therapy from Ecklonia cava. Phytother Res 2015; 29: 549-553
  • 43 Rana M, Pareek A, Bhardwaj S, Arya G, Nimesh S, Arya H, Bhatt TK, Yaragorla S, Sharma AK. Aryldiazoquinoline based multifunctional small molecules for modulating Aβ42 aggregation and cholinesterase activity related to Alzheimerʼs disease. RSC Adv 2020; 10: 28827-28837
  • 44 Koch N, Jennotte O, Gasparrini Y, Vandenbroucke F, Lechanteur A, Evrard B. Cannabidiol aqueous solubility enhancement: Comparison of three amorphous formulations strategies using different type of polymers. Int J Pharm 2020; 589: 119812
  • 45 Millar SA, Maguire RF, Yates AS, OʼSullivan SE. Towards better delivery of Cannabidiol (CBD). Pharmaceuticals (Basel) 2020; 13: 219
  • 46 Towler P, Staker B, Prasad SG, Menon S, Tang J, Parsons T, Ryan D, Fisher M, Williams D, Dales NA, Patane MA, Pantoliano MW. ACE2 X-ray structures reveal a large hinge-bending motion important for inhibitor binding and catalysis. J Biol Chem 2004; 279: 17996-18007
  • 47 Wang Q, Zhang Y, Wu L, Niu S, Song C, Zhang Z, Lu G, Qiao C, Hu Y, Yuen KY, Wang Q, Zhou H, Yan J, Qi J. Structural and functional basis of SARS-CoV-2 entry by using human ACE2. Cell 2020; 181: 894-904.e9
  • 48 Malin JJ, Suárez I, Priesner V, Fätkenheuer G, Rybniker J. Remdesivir against COVID-19 and other viral diseases. Clin Microbiol Rev 2020; 34: e00162-20
  • 49 Gordon CJ, Tchesnokov EP, Woolner E, Perry JK, Feng JY, Porter DP, Götte M. Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency. J Biol Chem 2020; 295: 6785-6797
  • 50 Maciorowski D, Idrissi SZE, Gupta Y, Medernach BJ, Burns MB, Becker DP, Durvasula R, Kempaiah P. A review of the preclinical and clinical efficacy of remdesivir, hydroxychloroquine, and lopinavir-ritonavir treatments against COVID-19. SLAS Discov 2020; 25: 1108-1122
  • 51 Carr NT. Using Microsoft Excel® to calculate descriptive statistics and create graphs. Lang Assess Q 2008; 5: 43-62
  • 52 Papautsky EL, Hamlish T. Patient-reported treatment delays in breast cancer care during the COVID-19 pandemic. Breast Cancer Res Treat 2020; 184: 249-254
  • 53 Becker RC. COVID-19 and its sequelae: a platform for optimal patient care, discovery and training. J Thromb Thrombolysis 2021;
  • 54 Jasper EE, Ajibola VO, Onwuka JC. Nonlinear regression analysis of the sorption of crystal violet and methylene blue from aqueous solutions onto an agro-waste derived activated carbon. Appl Water Sci 2020; 10: 132
  • 55 R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2021. Accessed March 16 2021 at: https://www.R-project.org/
  • 56 R Studio Team. RStudio: Integrated Development Environment for R. Boston, MA: RStudio, PBC; 2021. Accessed March 16, 2021 at: http://www.rstudio.com/
  • 57 Baty F, Ritz C, Charles S, Brutsche M, Flandrois JP, Delignette-Muller ML. A toolbox for nonlinear regression in R: The package nlstools. J Stat Softw 2015; 66: 1-21
  • 58 Zeviani WM. wzRfun: Walmes Zevianiʼs collection of functions. 2019. Accessed March 18, 2021 at: https://github.com/walmes/wzRfun
  • 59 Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, Hutchison GR. Avogadro: An advanced semantic chemical editor, visualization, and analysis platform. J Cheminformatics 2012; 4: 17
  • 60 Trott O, Olson AJ. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31: 455-461
  • 61 Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE. UCSF Chimera–A visualization system for exploratory research and analysis. J Comput Chem 2004; 25: 1605-1612
  • 62 Schneider N, Hindle S, Lange G, Klein R, Albrecht J, Briem H, Beyer K, Claußen H, Gastreich M, Lemmen C, Rarey M. Substantial improvements in large-scale redocking and screening using the novel HYDE scoring function. J Comput Aided Mol Des 2012; 26: 701-723
  • 63 Musoev A, Numonov S, You Z, Gao H. Discovery of novel DPP-IV inhibitors as potential candidates for the treatment of type 2 diabetes mellitus predicted by 3D QSAR Pharmacophore models, molecular docking and de novo evolution. Molecules 2019; 24: 2870
  • 64 Gurung AB, Ali MA, Lee J, Farah MA, Al-Anazi KM. Unravelling lead antiviral phytochemicals for the inhibition of SARS-CoV-2 Mpro enzyme through in silico approach. Life Sci 2020; 255: 117831
  • 65 Ramírez D, Caballero J. Is it reliable to take the molecular docking top scoring position as the best solution without considering available structural data?. Molecules 2018; 23: 1038
  • 66 Velázquez-Libera JL, Murillo-López JA, de la Torre AF, Caballero J. Structural requirements of N-alpha-Mercaptoacetyl Dipeptide (NAMdP) inhibitors of pseudomonas aeruginosa virulence factor LasB: 3D-QSAR, molecular docking, and interaction fingerprint studies. Int J Mol Sci 2019; 20: 6133
  • 67 Biovia DS. Discovery Studio Visualizer. San Diego, CA, USA: Dassault Systèmes; 2017: 936

Zoom Image
Fig. 1 Simplified replication cycle of coronaviruses in the human host cell: (1) indicates one of our targets, inhibiting hACE2 activity in which will prevent the coronaviruses to enter the human cell; (2) indicates the other target, inhibiting the viral main protease in which will stop the viral replication.
Zoom Image
Fig. 2 Percent inhibition of GC376 as the positive control, THC, CBD, and CBN against SARS-CoV1 and SARS-CoV2 main protease (MPro) activities.
Zoom Image
Fig. 3 Concentration-response curves of GC376 (a), CBD (b), and THC (c) against SARS-CoV2 main protease (MPro) present in normal scales.
Zoom Image
Fig. 4 Structure-activity relationship of THC, CBD, and CBN against SARS-CoV1/2 main protease (MPro) activities compare with the actual experiment of each cannabinoid highlighting important chemical features on the cannabinoid structure. The red circle indicates an essential component on ring B, while the green circle indicates free rotation as a key feature.
Zoom Image
Fig. 5 Detailed 3D pose and molecular interaction of THC, CBD, CBN, and GC376 and SARS-CoV2 main protease (PDB ID: 7C6U) including the involved amino acids: a The 3D binding pose of GC376 in the binding pocket of SARS-CoV2 main protease; b Superposition 3D structures of THC, CBD, CBN, and GC376 in the binding pocket of SARS-CoV2 main protease; c The 3D binding pose of CBD; d The 3D binding pose of THC; e the 3D binding pose of CBN and SARS-CoV2 main protease; f The 2D molecular diagram of CBD; g The 2D molecular diagram of THC; h The 2D molecular diagram of CBN and the involved amino acids in the binding pocket of SARS-CoV2 main protease; A, B, and C indicates the core chemical components on the cannabinoid structure; The green and red circle indicate the key chemical feature in CBD and CBN, respectively.
Zoom Image
Fig. 6 Concentration-response curves of MLN-4760 (a), CBD (b), and THC (c) against hACE2 enzyme present in normal scales.
Zoom Image
Fig. 7 Detailed 3D pose and molecular interaction of THC, CBD, MLN-4760, and hACE2 (PDB ID: 1R4L) including the involved amino acids: a The 3D binding pose of the hACE2-MLN-4760 complex (PDB ID: 1R4L, brown color) in comparison to the hACE2-SARS-CoV2 RBD complex (PDB ID: 6LZG, pink and blue color). The narrow indicates the changing of sub-domain I between native hACE2 and hACE2-inhibitor complex, approximately 13 Å, and AS indicates the active site; b The 3D binding pose of MLN-4760 in the binding pocket of hACE2. S1 and S1′ indicates the sub-pockets in the active site; c Superposition 3D structures of THC, CBD, and MLN-4760 in the binding pocket of hACE2; d 3D binding pose of CBD; e 3D binding pose of THC; f 2D molecular diagram of CBD; g 2D molecular diagram of THC and the involved amino acids in the binding pocket of hACE2; A, B, and C indicates the core chemical components on the cannabinoid structure.