Open Access
CC BY 4.0 · Pharmaceutical Fronts
DOI: 10.1055/a-2731-9183
Original Article

Virtual Screening of Anti-HIV Leads from Mayana (Coleus scutellarioides Benth.) Phytoconstituents

Authors

  • Von Jay Maico G. Gabucan

    1   College of Pharmacy and Chemistry, University of the Immaculate Conception, Davao City, Philippines

Funding None.
 

Abstract

While some Coleus species have demonstrated anti-HIV activity, the potential of Mayana (Coleus scutellarioides Benth.) remains largely unexplored. This study therefore aimed to investigate the anti-HIV potential of phytoconstituents from Mayana using in silico methods. Phytochemicals from Mayana were identified using gas chromatography–mass spectrometry and their binding affinity against HIV-1 integrase (IN), protease (PR), and reverse transcriptase (RT) were evaluated through molecular docking simulations. In this work, a total of 32 individual compounds were identified. Stigmasterol was found to have the highest binding affinity to HIV IN (−8.571 kcal/mol) and HIV PR (−10.250 kcal/mol), whereas caryophyllene showed the highest affinity to HIV RT (−9.625 kcal/mol). These compounds also demonstrated multitarget interactions, suggesting potential inhibitory effects. However, compared with synthetic drugs such as amprenavir (−9.421 kcal/mol for PR), raltegravir (−9.825 kcal/mol for IN), and nevirapine (−9.748 kcal/mol for RT), the phytoconstituents had lower binding affinities. Pharmacokinetic predictions revealed that the top-ranked phytochemicals conform to Lipinski's Rule of Five, indicating favorable drug-like properties. Overall, Mayana contains bioactive phytoconstituents with promising affinity for key HIV-1 enzymes, supporting the potential of Mayana as a source of novel anti-HIV leads. However, further in vitro and in vivo studies are needed to confirm the efficacy, safety, and pharmacokinetic profile.


Introduction

The human immunodeficiency virus (HIV) remains a major global health challenge, with millions affected worldwide. Despite significant advancements in antiretroviral therapy (ART), HIV remains incurable and requires lifelong treatment to suppress viral replication. Although ART effectively suppresses viral replication, its utility is compromised by drug resistance, adverse effects, and accessibility challenges, particularly in resource-limited settings. These issues highlight the need for alternative or complementary therapeutic strategies, particularly those derived from natural sources. As a result, some people turn to natural products and herbal medicines, which are often perceived as safer and having fewer undesirable effects than synthetic drugs.[1]

ART is associated with a range of adverse effects that can significantly impair patient adherence. Commonly reported side effects, including nausea, dizziness, fatigue, difficulty in concentration, and diarrhea, are significant factors influencing treatment compliance.[2] Other frequent complications encompass anemia, hepatotoxicity, hypertriglyceridemia, and peripheral neuropathy, as well as allergic reactions like skin rashes and itching.[3] Furthermore, the development of drug resistance to antiretroviral medications remains a critical concern, especially in patients on long-term regimens.[4] Although the prevalence of resistance has decreased with modern ART, its prevention is essential to ensure sustained treatment success and preserve future therapeutic options.

Herbal medicine and remedies are proven to be beneficial as a treatment for HIV infections. The use of an herbal remedy was proven effective in treating HIV infection by improving clinical features and laboratory results.[5] Also, herbal medicines were used to treat infections secondary to HIV, but can also be used to reduce the side effects of antiretroviral drugs and to provide an overall positive effect on well-being.[6] Although antiretroviral drugs produce a potent and immediate improvement in CD4 cell counts, Chinese herbal medicine produces a steady rise and can provide a higher CD4 cell count increase than drugs alone.[7] These studies highlight the contribution and benefits that herbal medicine may provide when considering herbal medicine as an option in the treatment of HIV.

Moreover, species from the Coleus genus have shown inhibitory effects against HIV, which is primarily attributed to diterpenes and polyphenolic compounds.[8] [9] [10] While certain members like Coleus forskohlii and Coleus parvifolius have been studied for their anti-HIV activity, research on Mayana (Coleus scutellarioides Benth.) remains limited. Mayana is widely available in the Philippines. Mayana is an ornamental plant that is abundant in the Philippines and has been reported to be ethnobotanically used as an herbal medicine for various conditions and diseases.[11] [12] [13] [14] [15] Given the documented anti-HIV activity in some Coleus species, Mayana may also possess similar bioactivity. However, published studies directly investigating its efficacy against HIV are scarce. Traditional experimental methods, which are often resource-intensive and time-consuming, further limit research in this area. To bridge this gap, computational approaches such as molecular docking offer a viable and efficient alternative.

Molecular docking is an in silico approach to virtually simulate interactions between two molecules.[16] This tool can be used to investigate the mechanisms underlying molecular recognition, interaction, and also to identify the conformation of a receptor–ligand complex.[17] It has been an integral tool for drug discovery, especially in lead identification. In natural product research, molecular docking has gained prominence as a computational tool that predicts interactions between phytochemicals and disease targets, providing valuable insights for further studies and natural product development.[18] This approach offers a more efficient alternative to traditional drug discovery processes, reducing attrition rates by prioritizing promising candidates before costly and time-consuming experimental validation.[18]

As such, this study aims to identify the phytoconstituents present in the crude extracts of Mayana leaves and explore the potential of these phytoconstituents against target HIV enzymes using molecular docking software and a virtual screening approach. By investigating Mayana's bioactive compounds, this study seeks to contribute to the growing field of plant-based drug discovery for HIV treatment and encourage further experimental validation.


Materials and Methods

Research Design

This study employs an exploratory laboratory experimental design to identify the bioactive phytochemicals present in Mayana. Furthermore, the potential of these phytochemicals to interact with major HIV-1 enzymes such as integrase (IN), protease (PR), and reverse transcriptase (RT) was also determined through an in silico approach. The input–process–output framework was used to systematically structure the workflow of this study, from the extraction and identification of phytochemicals to the molecular docking analysis.


Preparation and Extraction of Crude Extract

The dried and powdered Mayana leaves that were procured from Herbanext Laboratories, Inc., were portioned for maceration in two different solvents. A total of 100 g of the plant material was submerged in enough 95% ethanol, and another 100 g sample was submerged in 500 mL of n-hexane. These were set aside and allowed to macerate for 3 days with frequent shaking. Using both ethanol and hexane as solvents for polar and nonpolar compounds will maximize the phytochemicals that can be extracted from the plant material. After maceration, the marc was filtered out using vacuum filtration, then gravity filtration. The resulting polar and nonpolar extracts were concentrated using a rotary evaporator (temperature of the water bath at 60°C and pressure at 600 mm Hg) and were dried using a lyophilizer. The lyophilized powder was then subjected to gas chromatography–mass spectrometry (GC–MS) analysis.


Identification Using Gas Chromatography–Mass Spectrometry Analysis

Analysis using GC–MS was carried out at Ateneo De Davao University, Davao City, Philippines. The specific instrument model used was GC–MS-QP2010 ULTRA (Shimadzu, Japan). Column oven temperature was maintained at 60°C. Splitless injection mode was used with an injection temperature of 220°C. Helium was used as a courier gas with a pressure of 100 kPa, and the sampling time was 1 minute. The column used was a Rtx5MS capillary column with specifications of 30 m length with 0.25 mm internal diameter, and 0.25-µm film thickness. Ion source temperature was maintained at 300°C, whereas interface temperature was maintained at 290°C. The peaks obtained from the mass spectra were compared and validated with standard spectra available from NIST08 (National Institute of Standards and Technology, United States) and Wiley7 (Wiley Science Solutions, United States) databases. A peak quality below 80% was excluded from the study.


Virtual Screening Using Molecular Docking

Molecular docking was carried out using AutoDock Vina (Scripps Research Institute, United States). AutoDock Vina is one of the fastest and most widely used virtual screening tools that offers a more accurate prediction of binding mode.[19] [20] For a streamlined and flexible molecular docking workflow, Dockey was used. Dockey is an open-access software (downloadable at https://github.com/lmdu/dockey) that is cross-platform compatible, easy to use, flexible, and implements the whole molecular docking pipeline in one intuitive graphical interface and making it simple for all types of researchers.[21] The docking study and its preparations were performed on a 2022 MacBook Pro 13” (Apple, United States) with 512 gigabytes of internal storage and eight gigabytes of memory.

HIV-1 IN (PDB Code: 3OYA), PR (PDB Code: 3NU3), and RT (PDB Code: 3LP1) were obtained from RCSB Protein Data Bank (https://www.rcsb.org). The enzymes were prepared by removing water, solvent, organics, and heteroatom nonreceptor atoms of the respective protein enzymes through the Docking software. Receptor preparation was done using AutoDock Tools, which is already integrated with Docking. Furthermore, grid box assignments for the various HIV enzymes followed the specification set by the study of Zubair et al.[22]

The structured data files of the phytochemicals identified from the GC–MS analysis of Mayana crude extract were collected from PubChem (https://pubchem.ncbi.nlm.nih.gov). The native ligands of the target enzymes, as based on a previous study, were also included as a form of validation of the results.[22] These native ligands include amprenavir, raltegravir, and nevirapine for HIV PR, IN, and RT, respectively. Native ligand docking and redocking were done to ascertain the most accurate receptor preparation algorithm and docking parameters. These ligands were also prepared for docking using AutoDock Tools, similarly to receptor preparation.

In the assessment or virtual screening through Docking, default parameters for docking were used. These parameters have been well-tested and highly optimized.[23] These default parameters include Vina as the scoring function and exhaustiveness of eight. Minimum root mean square deviation was 1.00 Å, and the maximum number of binding modes to generate was nine. The result of docking was expressed in binding free energy, and the ligand with the lowest value or score for the respective enzymes was associated with better affinity to the target enzymes, and the ligand with the most promising potential for bioactivity. LigPlot+ version 2.2 was used to illustrate the ligand–receptor complex interaction.[24] Furthermore, the ligand with the lowest binding free energy value was also associated with the most promising ligand conformation for the potential enzyme inhibitory activity.


Pharmacokinetic Prediction

Following molecular docking, the absorption, distribution, metabolism, excretion (ADME) prediction was carried out using SwissADME. SwissADME is a free web tool that provides researchers with a pool of robust predictive tools that assess the physicochemical properties, pharmacokinetics, drug-likeness, and medicinal chemistry friendliness of compounds.[25] Furthermore, the BOILED-Egg method was also used to easily visualize gastrointestinal absorption and brain access, which are two crucial pharmacokinetic behaviors.[26]

In doing the pharmacokinetic prediction, only the top ten Mayana phytochemicals that exhibited the lowest binding free energy were used. The selection of these phytochemicals was based on their strong binding affinities toward the target HIV enzymes, which suggested a greater likelihood of biological activity. By narrowing the pool to the most promising candidates, the study ensured that the pharmacokinetic evaluation would focus on compounds with both favorable docking profiles and potential drug-like characteristics. This approach not only streamlines the analysis but also increases the relevance of the ADME predictions.



Results

Through chromatographic analysis of the crude extracts of Mayana, 40 phytochemicals were identified. Some of these phytochemicals were found in both the ethanol and hexane extracts. These phytochemicals are reported in [Table 1]. From this table, the three most abundant phytochemicals based on % peak area were (3.beta.,23E)-9,19-cyclolanost-23-ene-3,25-diol (35.22%), methyl commate C (17.14%), and phytol (16.84%) in the ethanolic portion. All the while, the three most abundant in the hexane portion were squalene or 2,6,10,14,18, 22-tetracosahexaene, 2,6,10,15,19,23-hexamethyl-, (all-E)- (27.54%), tetrapentacontane (11.67%), and stigmasterol (3.24%).

Table 1

Phytochemicals identified from the ethanolic and hexane crude extract of Mayana leaves

Number

PubChem Compound ID

Identified Compound

RT (min)

Peak area (%)

Peak height (%)

Peak quality (%)

Extracted from

1

6420801

Sulfurous acid, butyl nonyl ester

5.08

0.03

0.26

81

EtOH

2

140213

3-Ethyl-3-methylheptane

5.12

0.13

1.16

92

EtOHa

3

12389

Tetradecane

5.15

0.09

0.73

84

EtOH

4

519387

Undecane, 3,7-dimethyl-

5.41

0.02

0.26

89

EtOH

5

519404

Undecane, 3,5-dimethyl-

5.46

0.03

0.31

85

EtOHa

6

569301

Silane, cyclohexyldimethoxymethyl-

5.49

0.04

0.39

87

EtOHa

7

2537

Bicyclo[2.2.1]heptan-2-one, 1,7,7-trimethyl-, (1R)-

5.52

0.06

0.49

91

EtOH

8

6552009

endo-Borneol

5.68

0.47

2.03

91

EtOH

9

136810

Benzene, 1,3-bis(1,1-dimethylethyl)-

6.09

0.06

0.36

81

EtOHa

10

545627

Dodecane, 4,6-dimethyl-

6.21

0.43

3.47

94

EtOHa

11

300476

Nonane, 5-butyl-

6.31

0.06

0.44

87

EtOH

12

551397

Nonane, 5-methyl-5-propyl-

6.36

0.07

0.61

88

EtOHa

13

595370

1H-Cyclopropa[a]naphthalene, decahydro-1,1,3a-trimethyl-7-methylene-, [1aS-(1a.alpha.,3a.alpha.,7a.beta.,7b.alpha.)]-

6.85

0.15

0.75

83

EtOHa

14

5281515

Caryophyllene

7.48

0.19

1.06

95

EtOHa

15

1742210

Caryophyllene oxide

9.33

0.95

1.28

92

EtOH

16

11007

Hexadecane, 1-iodo-

10.43

0.20

0.34

81

EtOH

17

8181

Hexadecanoic acid, methyl ester

13.87

0.40

0.84

92

EtOH

18

5280435

Phytol

16.50

16.84

9.66

95

EtOH

19

573073

5.beta.-iodomethyl-1.beta.-isopropenyl-4.alpha.,5.alpha.-dimethyl, 6.beta.Bicyclo[4.3.0]nonane

24.51

1.74

1.57

84

EtOH

20

*

Methyl Commate C

24.96

17.14

13.75

90

EtOH

21

609236

1,4-Dimethyl-8-isopropylidenetricyclo[5.3.0.0(4,10)]decane

25.71

4.53

3.88

84

EtOH

22

612782

4,4,6a,6b,8a,11,11,14b-Octamethyl-1,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-octadecahydro-2H-picen-3-one

25.96

4.77

4.11

82

EtOH

23

5363281

(3.beta.,23E)-9,19-Cyclolanost-23-ene-3,25-diol

26.60

35.22

20.78

86

EtOH

24

545936

Nonane, 5-(2-methylpropyl)-

5.11

0.13

0.84

93

Hex

25

140213

3-Ethyl-3-methylheptane

5.15

0.06

0.39

90

Hex

26

519404

Undecane, 3,5-dimethyl-

5.46

0.06

0.34

87

Hex

27

569301

Silane, cyclohexyldimethoxymethyl-

5.49

0.09

0.41

86

Hex

28

136810

Benzene, 1,3-bis(1,1-dimethylethyl)-

6.08

0.01

0.09

85

Hex

29

11006

Hexadecane

6.21

0.46

2.69

93

Hex

30

12398

Heptadecane

6.27

0.08

0.51

91

Hex

31

545627

Dodecane, 4,6-dimethyl-

6.51

0.17

0.89

92

Hex

32

551397

Nonane, 5-methyl-5-propyl-

6.58

0.04

0.19

87

Hex

33

5374422

Acetic acid, 3-(2,2-dimethyl-6-methylene-cyclohexylidene)-1-methyl-butyl ester

6.70

0.04

0.23

80

Hex

34

595370

1H-Cyclopropa[a]naphthalene, decahydro-1,1,3a-trimethyl-7-methylene-, [1aS-(1a.alpha.,3a.alpha.,7a.beta.,7b.alpha)]-

6.85

0.09

0.39

84

Hex

35

5281515

Caryophyllene

7.47

0.04

0.19

90

Hex

36

11008

Dotriacontane

20.29

0.48

0.56

86

Hex

37

5280794

Stigmasterol

22.56

3.24

3.31

83

Hex

38

12412

Hexatriacontane

23.48

1.52

1.76

91

Hex

39

521846

Tetrapentacontane

24.88

11.67

8.42

93

Hex

40

638072

2,6,10,14,18, 22-Tetracosahexaene, 2,6,10,15,19,23-hexamethyl-, (all-E)-

25.96

27.54

23.63

95

Hex

Notes: * indicated a sourced from SpectraBase with the compound ID LPM8fPGaWYa. Phytochemicals detected in both the ethanol (EtOH) and hexane (Hex) extracts are indicated with a superscript (a), shown in the EtOH row where they are first listed. A total of 32 individual phytochemicals were identified.


Molecular docking analysis was performed to predict the interactions between the phytochemicals and the three major HIV enzymes. Results are reported in [Table 2]. For HIV IN and HIV PR, the phytochemical with the lowest binding free energy is stigmasterol (−8.571 kcal/mol for HIV IN and −10.250 kcal/mol for HIV RT). Caryophyllene was predicted to have the highest binding affinity to HIV RT among all phytochemicals (−9.625 kcal/mol).

Table 2

Binding free energy of identified Mayana phytochemicals against HIV-1 enzymes

No

Compound identified

HIV IN

HIV PR

HIV RT

1

Sulfurous acid, butyl nonyl ester

−4.889

−5.002

−6.306

2

3-Ethyl-3-methylheptane

−4.285

−4.603

−6.078

3

Tetradecane

−4.099

−4.877

−6.500

4

Undecane, 3,7-dimethyl-

−4.850

−5.073

−6.425

5

Undecane, 3,5-dimethyl-

−4.483

−4.874

−4.602

6

Silane, cyclohexyldimethoxymethyl-

−5.191

−5.397

−6.658

7

Bicyclo[2.2.1]heptan-2-one, 1,7,7-trimethyl-, (1R)-

−5.061

−5.899

−5.975

8

endo-Borneol

−5.035

−5.565

−5.769

9

Benzene, 1,3-bis(1,1-dimethylethyl)-

−5.95710

−6.8858

−8.8384

10

Dodecane, 4,6-dimethyl-

−4.477

−5.318

−7.44710

11

Nonane, 5-butyl-

−4.387

−4.777

−4.655

12

Nonane, 5-methyl-5-propyl-

−4.686

−4.809

−6.742

13

1H-Cyclopropa[a]naphthalene, decahydro-1,1,3a-trimethyl-7-methylene-, [1aS-(1a.alpha.,3a.alpha.,7a.beta.,7b.alpha.)]-

−6.4704

−6.8759

−9.5292

14

Caryophyllene

−6.3275

−7.0087

−9.6251

15

Caryophyllene oxide

−6.0778

−7.4235

−8.5607

16

Hexadecane, 1-iodo-

−4.288

−5.072

−4.414

17

Hexadecanoic acid, methyl ester

−4.710

−5.135

−4.181

18

Phytol

−5.832

−6.272

−5.280

19

5.beta.-Iodomethyl-1.beta.-isopropenyl-4.alpha.,5.alpha.-dimethyl, 6.beta.bicyclo[4.3.0]nonane

−5.636

−6.217

−5.399

20

1,4-Dimethyl-8-isopropylidenetricyclo[5.3.0.0(4,10)]decane

−6.0299

−7.1316

−7.9989

21

4,4,6a,6b,8a,11,11,14b-Octamethyl-1,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-octadecahydro-2H-picen-3-one

−7.6753

−7.5264

−8.8345

22

(3.beta.,23E)-9,19-Cyclolanost-23-ene-3,25-diol

−7.8922

−8.8052

−8.6536

23

Nonane, 5-(2-methylpropyl)-

−4.491

−4.889

−7.169

24

Silane, cyclohexyldimethoxymethyl-

−5.191

−5.397

−6.658

25

Hexadecane

−4.340

−5.029

−6.969

26

Heptadecane

−4.537

−5.279

−6.860

27

Acetic acid, 3-(2,2-dimethyl-6-methylene-cyclohexylidene)-1-methyl-butyl ester

−6.3166

−6.70410

−5.909

28

Dotriacontane

−5.077

−5.903

−6.784

29

Stigmasterol

−8.5711

−10.2501

−8.4508

30

Hexatriacontane

−5.099

−5.911

−6.916

31

Tetrapentacontane

−5.344

−6.188

−5.882

32

2,6,10,14,18,22-Tetracosahexaene, 2,6,10,15,19,23-hexamethyl-, (all-E)-

−6.1407

−7.7463

−9.3123

Note: Superscript numbers indicate the top 10 ligands with the highest binding affinities for each target enzyme. Native ligands have the following binding affinity: −9.825 kcal/mol for raltegravir against HIV integrase (IN), −9.421 kcal/mol for amprenavir against HIV protease (PR), and −9.748 kcal/mol for nevirapine against HIV reverse transcriptase (RT). The ten phytochemicals with the lowest binding free energy values for each enzyme are distinguished with subscripts 1 to 10.


In comparison to native ligands, raltegravir scored −9.825 kcal/mol for HIV IN, amprenavir scored −9.421 kcal/mol for HIV PR, and nevirapine scored −9.748 kcal/mol for HIV RT.

Comparison between the interactions of the phytochemicals with the lowest binding free energy and the native ligands is reported in [Table 3] and visualized in [Table 4]. Raltegravir and stigmasterol share similar interactions with HIV IN in that they both interact with the residues Arg-350, Leu-113, Pro-115, and Lys-219. Amprenavir and stigmasterol both interact with HIV PR, mainly with the residues Ala-28, Asp-30, Ile-84, Ile-47 (chain B), and Ile-50 (chain B). Lastly, nevirapine and caryophyllene interact with HIV RT through the residues Leu-100, Tyr-181, and Leu-234.

Table 3

Drug likelihood and ADME prediction of Mayana phytochemicals with high binding affinity to HIV enzymes

PubChem

CID

Identified Compound

MW

Lipinski violations

GI absorption

BBB permeant

Pgp substrate

CYP1A2 inhibitor

CYP2C19 inhibitor

CYP2C9 inhibitor

CYP2D6 inhibitor

CYP3A4 inhibitor

5280794

Stigmasterol

412.69

1

Low

No

No

No

No

Yes

No

No

5363281

(3.beta.,23E)-9,19-Cyclolanost-23-ene-3,25-diol

484.75

1

Low

No

No

No

No

No

No

No

612782

4,4,6a,6b,8a,11,11,14b-Octamethyl-1,4,4a,5,6,6a,6b,7,8,8a,9,10,11,12,12a,14,14a,14b-octadecahydro-2H-picen-3-one

424.70

1

Low

No

No

No

No

No

No

No

595370

1H-Cyclopropa[a]naphthalene, decahydro-1,1,3a-trimethyl-7-methylene-, [1aS-(1a.alpha.,3a.alpha.,7a.beta.,7b.alpha.)]-

204.35

1

Low

No

No

No

Yes

Yes

No

No

5281515

Caryophyllene

204.35

1

Low

No

No

No

Yes

Yes

No

No

5374422

Acetic acid, 3-(2,2-dimethyl-6-methylene-cyclohexylidene)-1-methyl-butyl ester

250.38

0

High

Yes

No

No

Yes

Yes

No

No

638072

2,6,10,14,18, 22-Tetracosahexaene, 2,6,10,15,19,23-hexamethyl-, (all-E)-

410.72

0

High

Yes

No

No

Yes

Yes

No

No

1742210

Caryophyllene oxide

220.35

0

High

Yes

No

No

Yes

Yes

No

No

609236

1,4-Dimethyl-8-isopropylidenetricyclo[5.3.0.0(4,10)]decane

204.35

1

Low

No

No

No

Yes

Yes

No

No

545627

Dodecane, 4,6-dimethyl-

198.39

1

Low

No

No

No

No

Yes

No

No

136810

Benzene, 1,3-bis(1,1-dimethylethyl)-

198.39

1

Low

No

No

No

No

Yes

No

No

Abbreviation: GI, gastrointestinal.


Note: Lipinski violations stand for the number of violations to the Lipinski druggability rule or rule of 5.


Table 4

Visualization of interactions between the native ligands and the top phytochemical for each HIV enzyme

Enzyme

Native ligand

Mayana phytochemical

HIV reverse transcriptase

HIV integrase

HIV protease

Pharmacokinetic prediction results are tabulated in [Table 3] and visualized through the BOILED-Egg method in [Fig. 1]. A total of 11 compounds were used and analyzed with the SwissADME and BOILED-Egg method. The 11 compounds ranged in molecular weight from 198.39 to 484.75 daltons. In terms of the violations of drug-likeness rules, 8 of the 11 compounds were found to have only 1 violation—they had poor solubility, resulting in low gastrointestinal absorption. Compounds acetic acid, 3-(2,2-dimethyl-6-methylene-cyclohexylidene)-1-methyl-butyl ester (CID: 5374422), 2,6,10,14,18, 22-Tetracosahexaene, 2,6,10,15,19,23-hexamethyl-, (all-E)- (CID: 638072), and caryophyllene oxide (CID: 1742210) were predicted to have blood–brain barrier permeability. The rest of the compounds did not have any violations of the Lipinski rules. None of the compounds was predicted to be p-glycoprotein substrates.

Zoom
Fig. 1 Boiled-egg diagram for the top Mayana phytochemicals against HIV enzyme. Ligands with a red outline indicate they are not p-glycoprotein substrates. Ligands are labeled using their PubChem CID. Three molecules were outside of range: 612782, 638072, 136810.

Discussion

This study used a molecular docking program to investigate the potential of Mayana and its constituents as agents against selected HIV-1 enzymes. These phytochemical constituents were identified from the ethanolic and hexane extracts of dried Mayana powders using GC–MS. Through molecular docking, the binding affinity of these phytochemicals was predicted and elaborated.

Through the chromatographic analysis, around 32 individual organic compounds from a variety of classes were identified. Upon closer examination, these compounds mostly consisted of the terpenoid family, steroids, and hydrocarbon compounds. These terpenoid compounds include monoterpenes, sesquiterpenes, diterpenes, and triterpenes. The findings align with phytochemical analyses of other studies, confirming the presence of terpenoid compounds, steroids, and steroid-based phytochemicals in Mayana.[27] [28] However, unlike other studies where flavonoids and phenols were qualitatively indicated, the current study did not show compounds of flavonoid or phenolic nature. This may be a result of drying since flavonoids and phenolic compounds have been known to degrade in heat.[29] [30] [31]

Results of docking analysis have also been reported in the literature. The binding free energy measures the stability of the complex formed between the ligand and the target protein.[32] The smaller the binding free energy, the more stable the complex is, and the higher the binding affinity is. The phytochemicals having the lowest binding free energy were caryophyllene for HIV RT (−9.625 kcal/mol) and stigmasterol for both HIV IN (−8.571 kcal/mol) and HIV PR (−10.25 kcal/mol). These compounds are terpenoid compounds, which align with previously conducted studies on the anti-HIV activity of terpenoids.[33] [34] [35]

Stigmasterol is one of the most common plant sterols found in nature and has been reported to have a variety of pharmacological properties, including anti-HIV activity.[36] [37] Another study has also reported that stigmasterol extracted from stem bark and leaf extracts of Kra Don (Careya arborea Roxb.) exhibited bioactivity against HIV RT with inhibition levels comparable to nevirapine.[38] Furthermore, the binding affinity of stigmasterol for HIV PR aligns with the findings of Zubair et al.[22] Stigmasterol was also reported to have inhibitory activity against HIV IN.[39]

Caryophyllene is a natural bicyclic sesquiterpene that possesses extensive biological activities.[40] Caryophyllene has also demonstrated preventive effects of mechanical allodynia induced by nucleoside RT in mouse models.[41] Many plants containing caryophyllene have also reported inhibitory effects on HIV-1 reverse transcriptase, with varying levels of activity.[42] Plant species containing caryophyllene and caryophyllene oxide were also reported to exhibit anti-HIV activity against HIV-infected lymphocyte cells.[43] Although the HIV-1 RT inhibitory activity of β-caryophyllene itself has not been validated in vitro, plant extracts containing this compound have demonstrated relevant bioactivity. The findings of this study provide an in silico basis that warrants further investigation on caryophyllene.

A comparison of binding affinities revealed that the native synthetic drugs generally outperformed the phytochemicals, except in the case of HIV PR, where certain plant-derived compounds showed superior or comparable binding. The binding free energies of raltegravir, amprenavir, and nevirapine were −9.825, −9.421, and −9.748 kcal/mol, respectively, indicating that the drugs remain superior. For amprenavir, the results are similar to the study of Zubair et al, where the plant metabolites had a lower free binding energy than the same native ligands.[22] These phytochemicals may have a higher affinity to HIV PR than amprenavir, implying better inhibitory activity. However, in-depth investigations are needed to confirm this claim.

An analysis of the underlying interactions reveals similar and overlapping binding modes between the compounds and the target enzymes. For HIV IN, both raltegravir and stigmasterol formed hydrogen bonds and hydrophobic interactions. These interactions span the essential domains of HIV IN, which are critical for enzymatic function.[44] Similarly, amprenavir and stigmasterol shared identical interaction types and residues in HIV PR. Notably, stigmasterol interacts with both Asp-25 residues in the dimeric HIV PR enzyme, a key component of the catalytic active site.[45] Furthermore, stigmasterol also interacts with the amino acid residues of the flap regions, which are important for securing substrate binding. Lastly, nevirapine and caryophyllene interact with amino acid residues of HIV RT through hydrophobic interactions. The amino acid residues with which caryophyllene interacts are the identical residues that RNase H inhibitors interact with,[46] suggesting it works by inhibiting RNase H, which is an important catalytic active site of HIV RT.[47]

The top phytochemicals in Mayana, especially stigmasterol, also showed similar affinity to the other two HIV enzymes. Some other Mayana compounds display affinities comparable to stigmasterol and caryophyllene. These phytochemicals may be beneficial due to their ability to target multiple enzymes, a key area of research interest.[48] [49] The findings support further research on Mayana as a plant-based therapy for HIV, although more studies, including the isolation of specific compounds and additional testing, are needed to confirm its anti-HIV effects.

For ADME prediction, the analysis revealed that the 11 phytochemicals predicted with the highest binding affinity to HIV target enzymes generally adhered to Lipinski's rule of 5, with at most one violation each. This suggests that they possess favorable drug-like properties. The Lipinski's rule of 5, also called the druggability rule, is a widely used “rule of thumb” for evaluating whether a substance has characteristics consistent with orally active drugs.[50] Caryophyllene oxide was predicted to have no violations with the Lipinski rules, but caryophyllene had one—it has poor solubility. This suggests that the structure of caryophyllene should be slightly modified, such as by introducing polar functional groups, to improve its pharmacokinetic properties without significantly compromising its bioactivity.

Sterol-like compounds such as stigmasterol and (3.beta.,23E)-9,19-cyclolanost-23-ene-3,25-diol showed strong binding affinities across HIV target enzymes. These two sterol compounds had binding free energy values that approached the active ligands closely. However, these compounds were found to have limitations with their drug-likeness as they were predicted to have low gastrointestinal absorption. This can be circumvented by chemical modifications or formulating these compounds in a suitable drug delivery system.

Taken together, docking and ADME predictions suggest that while sterol derivatives may serve as strong bioactive leads, compounds such as acetic acid ester, tetracosahexaene, and caryophyllene oxide emerge as the most balanced candidates, offering both significant enzyme inhibition potential and favorable drug-like properties for further development.

Limitations

Firstly, compounds must be sufficiently volatile to transfer from the liquid to the gas phase to be eluted from the analytical column.[51] Without adequate volatility, they cannot be eluted or detected. As no chemical derivatization was performed to facilitate the identification of flavonoids, this likely resulted in the absence of identified flavonoid and phenolic compounds in the analysis. Future studies should consider using high-performance liquid chromatography or other chromatographic techniques that better account for phytochemical stability. Second, the molecular docking findings of this research require validation through in vitro and in vivo studies. While molecular docking predicts binding potential, it does not confirm actual enzymatic inhibition. Furthermore, although this study focused on three key enzymes, phytochemicals may also interact indirectly with these enzymes or through other signaling pathways. Despite these limitations, the findings remain relevant and provide a valuable contribution to the field of natural product-based HIV drug discovery.



Conclusion

This virtual screening study provides in silico evidence that Mayana phytochemicals, particularly terpenoids, can target HIV IN, PR, and RT, supporting their great potential as anti-HIV agents. While their binding affinities are generally lower than those of synthetic drugs, these natural compounds represent promising lead structures for further optimization. Future work should focus on the targeted isolation of these specific compounds, followed by experimental validation of their bioactivity, safety, and efficacy through in vitro and in vivo studies.



Conflict of Interest

None declared.

Supporting Information

The original docking result for HIV integrase (IN), protease (PR), and reverse transcriptase (RT) ([Supplementary Tables S1]–[S3], available in online version) can be found in the “Supporting Information” section of this article's webpage.



Address for correspondence

Von Jay Maico G. Gabucan, RPh
College of Pharmacy and Chemistry, University of the Immaculate Conception
Davao City 8000
Philippines   

Publication History

Received: 21 March 2025

Accepted: 24 October 2025

Article published online:
25 November 2025

© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany


Zoom
Fig. 1 Boiled-egg diagram for the top Mayana phytochemicals against HIV enzyme. Ligands with a red outline indicate they are not p-glycoprotein substrates. Ligands are labeled using their PubChem CID. Three molecules were outside of range: 612782, 638072, 136810.