Next Article in Journal
Curcumin Alleviates Doxorubicin-Induced Cardiotoxicity by Modulating Apelin Expression
Previous Article in Journal
Red-Wine Gene Networks Linked to Exceptional Longevity in Humans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

UHPLC-QTOF-ESI-MS/MS, SNAP-MS Identification, In Silico Prediction of Pharmacokinetic Properties of Constituents from the Stem Bark of Holarrhena floribunda (G. Don) T. Durand and Schinz (Apocynaceae)

by
Franck Landry Djila Possi
1,2,
Mc Jesus Kinyok
2,
Joseph Eric Mbasso Tameko
2,
Bel Youssouf G. Mountessou
2,
Johanne Kevine Jumeta Dongmo
1,2,
Mariscal Brice Tchatat Tali
3,
Appolinaire Kene Dongmo
4,5,*,
Fabrice Fekam Boyom
3,
Jean Jules Kezetas Bankeu
2,
Norbert Sewald
4,
Jean Rodolphe Chouna
5 and
Bruno Ndjakou Lenta
2,4
1
Department of Organic Chemistry, Faculty of Science, University of Yaoundé, P.O. Box 812, Yaoundé 00001, Cameroon
2
Department of Chemistry, Higher Teacher Training College, University of Yaoundé, P.O. Box 47, Yaoundé 00001, Cameroon
3
Department of Biochemistry, Faculty of Science, University of Yaoundé, P.O. Box 812, Yaoundé 00001, Cameroon
4
Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, P.O. Box 100131, D-33501 Bielefeld, Germany
5
Department of Chemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 67, Cameroon
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(10), 1415; https://doi.org/10.3390/biom15101415
Submission received: 29 August 2025 / Revised: 26 September 2025 / Accepted: 29 September 2025 / Published: 4 October 2025
(This article belongs to the Section Natural and Bio-derived Molecules)

Abstract

The present work reports the bioguided isolation of constituents from the ethanol extract of Holarrhena floribunda stem bark, their identification by UHPLC-ESI-QTOF-MS/MS identification, and the in silico prediction of the pharmacokinetic and toxicity parameters. The crude extract, along with its n-hexane and alkaloid-rich fractions, displayed moderate to good antiplasmodial activity in vitro against chloroquine-sensitive (3D7) and multidrug-resistant (Dd2) strains of Plasmodium falciparum, with IC50 values ranging from 6.54 to 43.54 µg/mL. Seventeen steroidal alkaloids (117) were identified in the most active fraction using UHPLC-ESI-QTOF-MS/MS, based on their fragmentation patterns and analysis with the Structural Similarity Network Annotation Platform for Mass Spectrometry (SNAP-MS). Furthermore, bioguided isolation of the ethanol extract yielded twenty-one compounds (3, 5, 10, 1416, 1831), whose structures were elucidated by spectroscopic methods. Among them, compounds 5, 14, and 27 showed the highest potency against the two strains of P. falciparum, with IC50 values between 25.97 and 55.78 µM. In addition, the in silico prediction of pharmacokinetic parameters and drug-likeness using the SwissADME web tool indicated that most of the evaluated compounds (1, 35, and 1416) complied with Lipinski’s rule of five.

Graphical Abstract

1. Introduction

Malaria remains a global health challenge, mainly in sub-Saharan Africa, where this disease continues to kill thousands of people [1]. Hence, according to the 2024 World Health Organization (WHO) report, there were approximately 269 million cases of malaria affecting people from sub-Saharan countries, accounting for 94% of worldwide cases in 2023 [2]. In sub-Saharan Africa, this disease constitutes one of the major threats [3], especially in Cameroon, where about 3 million cases and more than 3800 deaths were reported in 2021 [4]. The first-line treatment options for this disease are based on the use of artemisinin-based combination therapies (ACTs), as recommended by WHO [5]. In addition, there are two existing malaria vaccines approved by the World Health Organization. Unfortunately, these vaccines have not shown satisfactory effectiveness (only 40% for RTS,S and around 75% for Matrix 21). These concerns, the spreading malaria parasite resistance to the first-line therapies in endemic settings, and the limited effectiveness of available vaccines highlight the urgent need for improved solutions to the malaria plague. Medicinal plants have historically played a central role in the treatment of human diseases, as evidenced by the success stories of quinine and artemisinin. The sub-Saharan African region carries the lion’s share of malaria burden, but is endowed with one of the richest plant biodiversity, from which novel chemical scaffolds can be discovered and developed as alternatives to the current chemotherapeutic agents. In this line, Holarrhena floribunda, a plant widely used in West and Central Africa for treating multiple diseases including malaria, dysentery, diarrhea, infertility, abdominal pains, nausea, indigestion, and diabetes [6,7,8] appears as a realistic starting material to search for novel antimalarial pharmacophores. Previous biological and chemical studies on this plant have revealed antibacterial [7], antiplasmodial [8,9,10], antitrypanosomal, cytotoxic [9,10], antioxidant [11], and antimycobacterial [12] activities and led to the isolation of triterpenoids [8], steroidal alkaloids [10], flavonoids [13], and diterpenoids [14].
Owing to the ethnopharmacological profile of H. floribunda and the sparse reports on the experimental validation of the antiplasmodial properties of its constituents [8,12], we undertook an antiplasmodial-guided investigation of H. floribunda ethanolic extract, followed by in silico prediction of the drug-likeness properties of selected constituents. Therefore, the present study reports the identification of seventeen steroidal alkaloids (117) from the alkaloid-rich fraction through Structural Similarity Network Annotation Platform for Mass Spectrometry (SNAP-MS), in combination with fragmentation patterns and data from the literature. In addition, the in silico prediction of pharmacokinetic parameters and target prediction of compounds (1, 3, 5, 10, 11 and, 1416) identified from the most active fraction are also reported herein. Also, the isolation of twenty-one compounds (3, 5, 11, 14, 16, and 1832), as well as the in vitro antiplasmodial and cytotoxic activities of the extract, fractions, and compounds from the stem bark of this plant, are also discussed herein.

2. Materials and Methods

2.1. Reagent and Materials

For this study, ethanol was used as an extraction solvent for the plant material. n-hexane, methylene chloride and n-butanol were used for partitioning. n-Hexane, cyclohexane, ethyl acetate and methanol were used as pure or binary mixtures at different polarities for the purification of compounds. Column chromatography was carried out with silica gel 230–400 mesh (Merck, Darmstadt, Germany), 70–230 mesh (Merck, Darmstadt, Germany) or Sephadex LH-20 (Sigma-Aldrich, Munich, Germany) as stationary phase. Thin-layer chromatography (TLC) was performed on Merck pre-coated silica gel (60 F254) aluminum foil (Merck, Darmstadt, Germany), and spots were visualized by inspection under a UV lamp operating at 254 or 365 nm and then by spraying with diluted sulfuric acid (20%) or with Dragendorff reagent before heating at about 100 °C. High-resolution mass spectra were recorded using a QTOF Compact Spectrometer (Bruker Corporation, Bremen, Germany). The 1H and 13C NMR spectra were recorded on Bruker Avance 600 (1H NMR, 600 MHz and 13C NMR, 150 MHz). Chemical shifts were reported in δ (ppm) using deuterated solvent as an internal standard, while coupling constants (J) were measured in Hz.

2.2. UHPLC-QTOF-ESI-MS/MS Analyses

High-resolution mass spectra were obtained with a QTOF Spectrometer (Bruker Corporation, Germany) equipped with an electrospray (ESI) source. The spectrometer was operated in the positive mode (mass range: 100–1500, with a scan rate of 1.00 Hz) with automatic gain control to provide high-accuracy mass measurements within 0.40 ppm deviation, using Na formate as calibrant. The following parameters were used for experiments: spray voltage of 4.5 kV and capillary temperature of 220 °C. Nitrogen was used as sheath gas (9 L/min). The MS/MS analyses were operated using a collision-induced dissociation method (CID) with collision energy of 45 eV. The spectrometer was coupled to an Ultimate 3000 (Thermo Fisher, Waltham, MA, USA) UHPLC system equipped with a LC-pump, a Diode Array Detector (DAD) (λm: 190–600 nm), an auto sampler (injection volume 100 μL) and an Accucore C-18 Reversed phase column (50 × 2.1 mm, 2.6 µm, 150 Å) in an oven temperature of 35 °C. The mobile phases consisted of water + 0.1% formic acid (A) and acetonitrile + 0.1% formic acid (B). Separation was carried out at 35 °C, at a flow rate of 0.4 mL/min, used with the following multistep linear gradient: 5% B isocratic for 10 min, 5–60% B for 22 min, 60% B isocratic for 3 min, 60–95% B for 2 min, 95% B isocratic for 1 min, 95–5% B for 1 min. The initial conditions were maintained for 1 min.

2.3. In Vitro Antiplasmodial Activity

Plasmodium falciparum Culture and Growth Inhibition Assay

P. falciparum 3D7 (Chloroquine-sensitive) and Dd2 (multidrug-resistant) strains were obtained from the Biodefense and Emerging Infections (BEI) Research Resources (Manassas, VA, USA) and were maintained using a slightly modified Trager and Jensen method (2005) [15]. In fact, parasites were cultured in fresh O+ human red blood cells at 3% (v/v) hematocrit in RPMI 1640 culture media containing glutamax NaHCO3 (Gibco, Paisley, UK), supplemented with 25 mM HEPES (Gibco, UK), 1X hypoxanthine (Gibco, Grand Island, NY, USA), 20 µg/mL gentamicin (Gibco, Shanghai, China), and 0.5% Albumax II (Gibco, Invitrogen, New York, NY, USA). Prior to the drug efficacy test, parasites were synchronized at the ring stage by sorbitol treatment and cultured through one cycle (48 h).
Stock solutions (100 mg/mL (Extracts and fractions) and 10 mg/mL (compounds) were prepared in 100% DMSO and serially diluted (5-fold) in complete RPMI 1640 prior to activity studies. The diluted intermediate solutions were co-cultured with parasite cultures (1% parasitemia and 2% hematocrit) in 96-well plates to a final drug concentration ranging from 50 to 0.08 µM for compounds and 100 to 0.16 µg/mL for extracts and fractions. The final dimethylsulfoxide (DMSO) concentration per 100 μL culture per well was less than 0.5% for all the tested samples. Chloroquine and artemisinin at a range of 1–0.0016 µM each were used as a negative growth control (positive test control), while the solvent-treated culture (<0.5% DMSO) was used as a positive growth control (negative test control). Following 72 h of incubation at 37 °C in a 5% CO2 incubator, parasite growth was assessed by a SYBR green I-based DNA quantification assay. A total of 100 μL of SYBR Green I buffer {[6 μL of 10,000 × SYBR Green I (Invitrogen) + 600 µL of red blood cells lysis buffer [Tris (25 mM; pH 7.5)] + 360 µL of EDTA (7.5 mM) + 19.2 µL of parasites lysis solution [saponin (0.012%; w/v)] and 28.8 µL of Triton X − 100 (0.08%; v/v)]} was added to each well and incubated in the dark at 37 °C for 1 h. Fluorescence was measured using an infinite M200 plate reader with excitation and emission wavelengths at 485 and 538 nm, respectively. Mean half-maximal inhibitory concentrations (IC50 values) were derived by plotting percent growth against log drug concentration and fitting the response data to a variable slope sigmoidal curve function using GraphPad Prism v8.0. The antiplasmodial assays were performed in triplicate.

2.4. Plant Material

The stem bark of H. floribunda was collected in Kon-Yambetta (GPS coordinate: latitude: 4.83333, longitude: 11.06674°49′60″ N, 11°4′0″ E) in Mbam et Inoubou Division, Centre region of Cameroon, in April 2022. The sample was identified by comparison with an available sample recorded under the voucher specimen N° 49821 HNC by Mr. Nana Victor, a botanist of the National Herbarium of Cameroon.

Extraction and Isolation

The stem bark of H. floribunda was chopped into small pieces, air dried and ground to give 4.5 kg of fine powder. The powder was macerated at room temperature (about 28 °C) using ethanol and freed from solvent to afford 155.2 g of crude extract. The crude extract (145.6 g) was dissolved in 500 mL of acidified water (HCl, 3 < pH < 4) and subjected to the liquid–liquid partition, using n-hexane (1 L × 3) and methylene chloride (1 L × 3) to give n-hexane (50.6 g), methylene chloride (9.7 g) fractions and the acidic aqueous phase. The acidic aqueous phase was then basified using 25% ammonia solution (NH4OH) (9 < pH < 10) and partitioned with methylene chloride (1 L × 3) to give 24.5 g of alkaloid-rich fraction. The n-hexane fraction (49.0 g) was subjected to column chromatography and eluted with the mixture of n-hexane/CH2Cl2 of increasing polarity [(9:1–7:3, v/v), CH2Cl2/MeOH (10:0–0:10, v/v)] to afford three main subfractions F1–F3. Subfraction F1 (20.3 g) underwent series of column chromatography eluted with n-hexane/EtOAc of increasing polarity (9:1–10:0, v/v) to yield compounds 18 (100.2 mg, n-hexane/EtOAc, 9.5:0.5, v/v), 21 (15.2 mg, n-hexane/EtOAc, 9:1, v/v), and the mixture of compounds 19, and 20 (40.3 mg, n-hexane/EtOAc, 8.5:1.5, v/v). The purification of the subfraction F2 (14.7 g) with a silica gel column yielded compounds 26 (7.3 mg, n-hexane/EtOAc, 9:1, v/v), 27 (5.2 mg, n-hexane/EtOAc, 8:2, v/v), and 30 (103.0 mg, n-hexane/EtOAc, 7:3, v/v). F3 was a complex mixture and was not studied.
In addition, the CH2Cl2 fraction (9.7 g) was subjected to CC over silica gel and eluted with cyclohexane/EtOAc (8:2–2:8, v/v), EtOAc/MeOH (10:0–0:10) of increasing polarities to give 150 fractions (with a collection volume of 200 mL per fraction). These fractions were gathered based on their TLC profiles to give 3 main subfractions (G1–G3). The silica gel column chromatography of the subfraction G1 (2.5 g) gave a mixture of compounds 24 and 25 (50 mg, n-hexane/EtOAc, 7.5:2.5, v/v) and compound 22 (15.2 mg, n-hexane/EtOAc, 7:3, v/v). Furthermore, subfraction G2 (1.5 g) was subjected to CC over silica to afford compound 23 (10.1 mg, n-hexane/EtOAc, 6:4, v/v). The subfraction G3 (3.1 g) was subjected over silica gel and eluted with cyclohexane/EtOAc by the gradient of increasing polarity (8:2–5.5:4.5, v/v) to afford compound 32 (150.0 mg, cyclohexane/EtOAc, 7.5:3.5, v/v), and subfractions G31 (50.0 mg) and G32 (25.5 mg). Subfraction G31 (50.0 mg) eluted with cyclohexane/EtOAc of increasing polarities (7.5:2.5–3:7, v/v) led to isolation of compounds 31 (10.5 mg) and 26 (7.3 mg). The subfraction G32 (25.2 mg) was subjected to CC over Sephadex LH-20 to yield compound 29 (10.2 mg).
The CH2Cl2 alkaloid-rich fraction (22.0 g) was subjected to silica gel column chromatography and eluted with a gradient of increasing polarity with CH2Cl2/acetone/NH4OH (100:0:0.1–0:100:0.1, v/v/v) to give four subfractions (ALK1–ALK4). Subfraction ALK1 (2.1 g) was subjected to column chromatography and eluted with CH2Cl2/MeOH/NH4OH of increasing polarity (98:2:0.1–40:60:0.1, v/v/v) to afford compounds 14 (5.3 mg, 40:60, v/v) and 15 (4.8 mg, 98:2, v/v). ALK2 (4.2 g) was subjected to silica gel column and eluted with the mixture of acetone/MeOH/NH4OH (9:1:0.1–8.5:1.5:0.1, v/v/v) to give three sub-fractions (ALK21-ALK23). The sub-fraction ALK21 (0.4 g) was subjected to CC over Sephadex LH-20 gel and eluted with MeOH/H2O (85:15, v/v) to afford compounds 5 (12.3 mg) and 11 (4.8 mg). ALK3 (6.4 g) was purified through CC over silica gel to give two subfractions ALK31 and ALK32. A silica gel column chromatography of ALKA31 (0.95 g) afforded compounds 3 (5.0 mg, EtOAc/MeOH/NH4OH, 9:1:0.1, v/v/v) and 16 (3.4 mg, EtOAc/MeOH/NH4OH, 8.5:1.5:0.1, v/v/v). ALK4 was not studied because of its complexity.

2.5. UHPLC/MS/MS Data Processing

Mass spectrometry data were processed using Mzmine (version 4.3.0) and MSConvert (version 3.0.24267-b37d261) prior to uploading into the Structural Similarity Network Annotation Platform for Mass Spectrometry (SNAP-MS) database, available at www.npatlas.org/discover/snapms (accessed on 5 January 2025). The SNAP-MS parameters were set as follows: reference database, COCONUT; adduct, [M + H]+; mass error tolerance, 10 ppm; maximum cluster size, 5000; minimum compound group size, 3; minimum NPAtlas annotation cluster size, 3; and minimum result edges, 10,000. The analysis generated a Cytoscape-compatible file, which was visualized using Cytoscape (version 3.10.2) to obtain the molecular network. Filtering of the SNAP-MS network was performed using Sirius (version 5.8.6) and by comparison with the literature data.

2.6. SwissADME, SwissTarget Prediction and In Silico Oral Toxicity

In Silico prediction of pharmacokinetic and drug-likeness parameters remains very useful in drug discovery, as it accelerates development, reduces costs, minimizes risks, and helps prioritize promising candidates [16]. In this study, pharmacokinetic properties and drug-likeness were evaluated using SwissADME web tool (http://www.swissadme.ch/index.php, accessed on 20 March 2025). The target prediction of compounds that fulfilled drug-likeness criteria with favorable pharmacokinetic profiles was performed using the SwissTargetPrediction platform (http://www.swisstargetprediction.ch/ accessed on 20 March 2025). In silico oral toxicity was assessed using the ProTox-II tool (https://tox.charite.de/protox3/ accessed on 13 June 2025).

3. Results and Discussion

3.1. UHPLC-ESI-QTOF-MS/MS Identification of Compounds

The alkaloid-rich fraction of the stem bark of H. floribunda was analyzed by UHPLC-ESI-Q-TOF-MS/MS (Figure 1). Peaks absent from the base peak chromatogram (BPC) of the alkaloid-rich fraction were extracted and subjected to MS/MS fragmentation (Figure 1). Seventeen steroidal alkaloids (117) were identified by interpreting their fragmentation pathways (see Supporting Information), in comparison with the literature data and with the aid of a freely accessible SNAP-MS platform (www.npatlas.org/discover/snapms, accessed on 28 August 2025), which generated the corresponding subnetworks (Figure 2). The identified steroidal alkaloids were classified into two subclasses including the conanine-type [isoconessimine (1), regholarrhenine D (2), irheline (3), conimin/conamine (4), conessine (5), conessimine (6), 7α-hydroxylconessine (7), regholarrhenine E (8), holarrhetine (10), holarrhesine (11), solanopubamide B (12), and holarrhetine isomer (13)] and the pregnene-type [salignemamide D (9), holaphyllamine (14), holaphyllaminol (15), N-methylholaphyllamine (16), and Salignemamide D isomer (17)] (Table 1, Figure 3).

3.2. Isolation of Compounds

In a view to further confirm the chemical composition of the active alkaloid-rich fraction, it underwent a series of column chromatography over silica gel and Sephadex LH-20 to afford six steroidal alkaloids, including irheline (3) [18], conessine (5) [17], holarhesine (11) [17], holaphyllamine (14) [10], holaphyllaminol (15) [18], and the mixture of N-methylholaphyllamine (16) [18] and irheline (3) [18] (Figure 4).
In addition, the n-hexane fraction displayed moderate in vitro antiplasmodial activity against the chloroquine-sensitive and multi-resistant strains of Plasmodium falciparum 3D7 (Pf3D7) and Dd2 (PfDd2), respectively. This fraction was purified by a series of column chromatography over silica gel and Sephadex LH-20 to afford lupeol (21) [26] and α-amyrin acetate (18) [27], a mixture of stigmasterol (20) and β-sitosterol (19) [28], ethylorsalinate (26) [29], the aliphatic lactone 3-β-hydroxyicosan-1,5-β-olide (30) [30], and lichexanthone (27) [31] (Figure 4).
The CH2Cl2 fraction was inactive against the chloroquine-sensitive (3D7) and multi-resistant (Dd2) strains of P. falciparum. Nevertheless, in a view to contribute to the chemotaxonomic knowledge on the plant, this fraction was purified investigated and led to the isolation of betulinic acid (22) [32], platanic acid (23) [33], oleanolic acid (24) [34], ursolic acid (25) [35], lupeol 3,5-dihydroxyeicosanoate (31) [36], cycloart-23Z-en-3β,25-diol (32) [37], β-sitosterol-3-O-β-D-glucopyranosyl (28) [28], and scopoletin (29) [38] (Figure 4).

3.3. Chemotaxonomic Significance

To the best of our knowledge, ethylorsalinate (26) and 3-β-hydroxyicosan-1,5-β-olide (30) were isolated for the first time from the Apocynaceae family. Lichexanthone (27), scopoletin (29), and platanic acid (23) are reported in this genus for the first time. However, they were already isolated from other plants of the Apocynaceae family. Oleanolic acid and ursolic acid were already reported in H. curtisii King and Gamble [27], while conessine (5), holarhesine (11), holaphyllamine (14), and N-methylholaphyllamine (16) were also reported in H. africana [10]. Irehline (3) and holaphyllaminol (15) have been reported in H. pubescens [18]. Among the putatively identified compounds, 9 and 12 are reported for the first time in the Apocynaceae family. Nevertheless, they have been reported in the Buxaceae and Solanaceae families [23,25]. The other herein identified steroidal alkaloids (116) were already reported in the genus Holarrhena. Thus, the present work stands as a significant contribution to the chemotaxonomic knowledge on H. floribunda.

3.4. Biological Contribution

As far as the in vitro antiplasmodial activity is concerned, the crude ethanol extract of the stem bark of H. floribunda was tested in vitro and showed good to moderate activity toward the Pf3D7 and PfDd2 strains with IC50 values of 14.12 ± 3.10 and 23.32 ± 2.39 µg/mL, respectively [39]. The n-hexane soluble fraction from the crude ethanol extract showed moderate activity against the Pf3D7 and PfDd2 with IC50 values of 36.40 ± 1.60 and 43.54 ± 2.74 µg/mL, respectively [39]. The methylene chloride soluble fraction from the crude extract was inactive (IC50 > 100 µg/mL) against both strains. The alkaloid-rich fraction showed good in vitro antiplasmodial activity, with IC50 values of 6.55 ± 0.60 and 8.27 ± 0.70 µg/mL against Pf3D7 and PfDd2, respectively (Table 2) [39]. These results could be justified by the antagonistic effect and/or the low concentration of some constituents in the ethanol crude extract of the stem bark of H. floribunda. The in vitro antiplasmodial activity of the alkaloid-rich fraction of this plant is reported herein for the first time, to the best of our knowledge. Even though all the fractions did not display an antiplasmodial activity against the assessed strain of Plasmodium, the antiplasmodial potentials of H. floribunda extracts were reported by Fotie and collaborators in 2006 against PfD6 and PfW2.
Conessine (5) showed moderate activity against the Pf3D7 and PfDd2 strains with IC50 values of 25.97 and 28.51 µM, respectively. In addition, amongst the assessed compounds, lichexanthone (27) showed moderate activity towards the aforementioned strains with IC50 values of 33.41 and 36.46 µM, respectively. Furthermore, holaphyllamine (14) showed marginal activity with an IC50 value of 49.53 and 55.78 µM on Pf3D7 and PfDd2, respectively. However, compound 14 exhibited good cytotoxicity against the human cervix carcinoma KB-3-1 cancer cell lines with an IC50 value of 9.8 µM as compared to cryptophycin (IC50 = 1.3 × 10−5 μM), according to the National Cancer Institute [40].
The in vitro antiplasmodial activity of some identified and isolated steroids (1, 35, 10, 11, 15, and 16) against Plasmodium falciparum FCB1 and K1 strains had been reported in previous studies. Indeed, these compounds showed good in vitro antiplasmodial activity, with IC50 values ranging from 0.97 to 11.7 µM against the chloroquine-resistant FCB1 and multi-resistant K1 strains of P. falciparum, respectively (Table 3) [17,18,41].

3.5. In Silico Pharmacokinetic Parameters, Drug-likeness Properties and Targets Prediction of Some Identified Compounds

Given the promising in vitro antiplasmodial activity of the alkaloid-rich fraction from the stem bark extract of H. floribunda, selected alkaloids identified or isolated (1, 3, 4, 5, 10, 11, 14, 15, and 16) were subjected to computational analysis using the SwissADME predictor to evaluate their physicochemical, pharmacokinetic, and drug-likeness properties (Table 4). Target prediction was also performed for compounds fulfilling drug-likeness criteria using the SwissTarget predictor. The analysis revealed that all selected compounds displayed high gastrointestinal absorption and a bioavailability score of 0.55, in agreement with previously reported bioavailability data [42]. Most of the compounds were moderately or poorly soluble in water. Amongst the tested compounds, only compound 14 complied fully with Lipinski’s rule of five, while all compounds satisfied Veber’s and Egan’s rules. Overall, compound 14 emerged as the most promising candidate, showing no Lipinski violations and favorable predicted pharmacokinetic parameters. Among the compounds evaluated in the in silico study, several, including conessine (5), holarrhesine (10), hollaphyllamine (14), and N-methylholaphyllamine (16), were predicted to exhibit good brain penetration. Moreover, all the tested compounds showed high gastrointestinal absorption.
To assess the potential interaction of the compounds with protein targets, those fulfilling drug-likeness criteria (compounds 1, 3, 4, 5, 14, and 16) were subjected to target prediction using the SwissTarget Predictor. The prediction results, illustrated in the pie chart (Figure 5), indicated that compound 1 was most likely to inhibit enzymes (13.3%), family A G protein-coupled receptors (40%), and cytochrome P450 enzymes (6.7%). Similarly, compound 3 was predicted to inhibit enzymes (13.3%), family A G protein-coupled receptors (46.7%), and cytochrome P450 enzymes (13.3%). Compound 4 was predicted to inhibit enzymes (26.7%), family A G protein-coupled receptors (26.7%), and cytochrome P450 enzymes (6.7%). Also, compound 15 was predicted to inhibit cytochrome P450 enzymes (26.7%), hydrolases (6.7%), family A G protein-coupled receptors (13.3%), and enzymes (20%). Compound 14 was predicted to inhibit cytochrome P450 enzymes (13.3%), family A G protein-coupled receptors (26.7%), enzymes (20%), oxidoreductases (13.3%), and nuclear receptors (13.3%). Compound 16 was predicted to inhibit cytochrome P450 enzymes (13.3%), family A G protein-coupled receptors (40%), proteases (13.3%), and oxidoreductases (13.3%) (Figure 5). Most of the selected compounds are not substrates for P-pg and CYP (Cytochromes P-450 isoenzymes).

3.6. In Silico Oral Toxicity Prediction

The toxicity profile of a compound is a critical parameter in drug discovery [43]. Compounds 5, 14, and 27, which showed the highest activity against P. falciparum, were further subjected to an in silico toxicity evaluation to assess their safety. Compound 27 was predicted to be the least toxic, with an LD50 of 3200 mg/kg and classified as toxicity class V [44]. Compounds 5 and 27 exhibited LD50 values of 750 and 390 mg/kg, respectively, and were classified as toxicity class IV [44]. In addition, compounds 5 and 14 were predicted to be devoid of hepatotoxicity, nephrotoxicity, and cardiotoxicity (Table 5), although they showed potential respiratory toxicity with prediction scores ranging from 0.73 to 0.77. Similarly, compound 27 was predicted to lack hepatotoxicity but to display potential nephrotoxicity, cardiotoxicity, and respiratory toxicity (Table 5).

4. Conclusions

In summary, this study adopted a bioguided approach to identify antiplasmodial pharmacophores, which could serve as starting points for the development of novel antimalarial agents. The results highlight the promising activity of certain fractions of H. floribunda against both chloroquine-sensitive and multidrug-resistant strains of P. falciparum. Chemical analysis of this plant enabled detailed characterization of its alkaloid-rich fraction, revealing a diverse group of steroidal alkaloids with favorable drug-like properties as predicted by in silico assessments. The isolation of key active compounds further underscores the therapeutic potential inherent in the stem bark. Importantly, the observed safety margins suggest that these compounds have promise for further development. Overall, this work advances our understanding of the chemical constituents responsible for the plant’s traditional use in malaria treatment and supports its potential as a source for future antimalarial drug development.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom15101415/s1, Figures S1–S37: 1H and 13C NMR spectra of compounds 3, 5, 11, 1416 and 1832; Figure S38: Base Peak Chromatogram (BPC) of the alkaloid fraction from the stem bark of H. floribunda; Figures S39–S55: MS/MS spectra of compounds 1, 2, 411 and 1416; Scheme S1–S14: MS/MS fragmentation of compounds 1, 2, 414, and 16.

Author Contributions

Conceptualization, F.L.D.P.; J.E.M.T.; J.J.K.B. and M.J.K.; methodology, F.L.D.P.; A.K.D.; M.J.K. and B.Y.G.M.; software, F.L.D.P. and A.K.D.; validation, J.J.K.B.; J.R.C.; N.S. and B.N.L.; formal analysis, A.K.D. and J.K.J.D.; investigation, F.L.D.P.; M.B.T.T.; J.E.M.T.; J.K.J.D. and F.F.B.:, resources, B.N.L. and J.R.C.; data curation, A.K.D. and M.B.T.T.; writing—original draft preparation, F.L.D.P.; writing—review and editing, A.K.D.; B.Y.G.M.; F.F.B. and J.J.K.B.; visualization, F.L.D.P.; supervision, J.R.C. and B.N.L.; project administration, N.S. and B.N.L.; funding acquisition, N.S. and B.N.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the German Academic Exchange Service (DAAD) through the Yaoundé-Bielefeld Graduate School of Natural Products with Antiparasite and Antibacterial activities (YaBiNaPA), project N° 57561808, grant number 57561808. The German Academic Exchange Service (DAAD) through the Yaoundé-Bielefeld Graduate School of Natural Products with Antiparasite and Antibacterial activities (YaBiNaPA), project no 57561808 supported this work.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

The authors are grateful to the German Academic Exchange Service (DAAD) for the financial support granted to the Yaoundé-Bielefeld Graduate School of Natural Products with Antiparasite and Antibacterial activities (YaBiNaPA), project N° 57561808. B.N.L. is equally grateful to Alexander von Humboldt for the research stay at Bielefeld University.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
TLCThin Layer Chromatography
UHPLCUltra-High Performance Liquid Chromatography
QTOF-ESI-MS/MSQuadrupole Time Of Flight Electrospray Ionization Tandem Mass Spectrometry
SNAP-MSStructural Similarity Network Annotation Platform for Mass Spectrometry
ADMETAbsorption, Desorption, Metabolism, Excretion and Toxicity

References

  1. WHO. World Malaria Report; WHO: Geneva, Switzerland, 2023. [Google Scholar]
  2. WHO. World Malaria Report: Addressing Inequity in the Global Malaria Response; WHO: Geneva, Switzerland, 2024. [Google Scholar]
  3. El-Moamly, A.A. How can we get malaria control back on track? BMJ 2024, 385, q1408. [Google Scholar] [CrossRef]
  4. Cameroon Kicks Off Malaria Vaccine Rollout. Available online: https://www.afro.who.int/countries/cameroon/news/cameroon-kicks-malaria-vaccine-rollout (accessed on 18 August 2025).
  5. WHO. World Malaria Report; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  6. Gnangoran, B.N.; N’guessan, B.B.; Amoateng, P.; Dosso, K.; Yapo, A.P.; Ehile, E.E. Hypoglycaemic activity of ethanolic leaf extract and fractions of Holarrhena floribunda (Apocynaceae). J. Med. Biomed. Sci. 2012, 1, 46–54. [Google Scholar]
  7. Bogne, K.P.; Penlap, B.V.; Lontsi, D.; Etoa, F.X. Antibacterial activities of the extracts and conessine from Holarrhena floribunda G. Don. (Apocynaceae). Afr. J. Tradit. Complement. Altern. Med. 2008, 4, 352–356. [Google Scholar] [CrossRef] [PubMed]
  8. Fotie, J.; Bohle, D.S.; Leimanis, M.L.; Georges, E.; Rukunga, G.; Nkengfack, A.E. Lupeol long-chain fatty acid esters with antimalarial activity from Holarrhena floribunda. J. Nat. Prod. 2006, 69, 62–67. [Google Scholar] [CrossRef] [PubMed]
  9. Hoekou, Y.P.; Tchacondo, T.; Karou, S.D.; Yerbanga, R.S.; Achoribo, E.; Da, O.; Atakpama, W.; Batawila, K. Therapeutic potentials of ethanolic extract of leaves of Holarrhena floribunda (G. Don) Dur. and schinz (Apocynaceae). Afr. J. Tradit. Complement. Altern. Med. 2017, 14, 227–233. [Google Scholar] [CrossRef]
  10. Nnadi, C.O.; Nwodo, N.J.; Kaiser, M.; Brun, R.; Schmidt, T.J. Steroid alkaloids from Holarrhena africana with strong activity against Trypanosoma brucei rhodesiense. Molecules 2017, 22, 1129. [Google Scholar] [CrossRef]
  11. Badmus, J.A.; Ekpo, O.E.; Hussein, A.A.; Meyer, M.; His, D.C. Antiproliferative and apoptosis induction potential of the methanolic leaf extract of Holarrhena floribunda (G. Don). Evid. Based Complement. Alternat. Med. 2015, 2015, 756482. [Google Scholar] [CrossRef]
  12. Yemoa, A.; Gbenou, J.; Affolabi, D.M.; Moudachirou, M.; Bigot, A.; Anagonou, S.; Portaels, F.; Martin, A.; Quetin-Leclercq, J. Beninese medicinal plants as a source of antimycobacterial agents: Bioguided fractionation and in vitro activity of alkaloids isolated from Holarrhena floribunda used in traditional treatment of buruli ulcer. Biomed Res. Int. 2015, 2015, 835767. [Google Scholar] [CrossRef]
  13. Badmus, J.A.; Okobi, E.; Rautenbach, F.; Marnewick, J.L.; Hussein, A.; Hiss, D.C. Isolation and antioxidant activity of flavonoids from Holarrhena floribunda (G.don) leaves. Acta Biochim. Pol. 2016, 63, 353–358. [Google Scholar] [CrossRef]
  14. Loukaci, A.; Kayser, O.; Bindseil, K.U.; Siems, K.; Frevert, J.; Abreu, M.P. New trichothecenes isolated from Holarrhena floribunda. J. Nat. Prod. 2000, 63, 52–56. [Google Scholar] [CrossRef]
  15. Trager, W.; Jensen, J.B. Human malaria parasites in continuous culture. J. Parasitol. 2005, 91, 484–486. [Google Scholar] [CrossRef]
  16. Alshammari, F. Application of computational tools for ADME and target modeling of bioactive compounds from hydroalcoholic extracts of Erodium glaucophyllum flowers. Med. Sci. 2020, 24, 4178–4183. [Google Scholar]
  17. Zirihi, G.N.; Grellier, P.; Guédé-Guina, F.; Bodo, B.; Mambu, L. Isolation, characterization and antiplasmodial activity of steroidal alkaloids from Funtumia elastica (Preuss) Stapf. Bioorg. Med. Chem. Lett. 2005, 15, 2637–2640. [Google Scholar] [CrossRef]
  18. Cheenpracha, S.; Boapun, P.; Limtharakul Née Ritthiwigrom, T.; Laphookhieo, S.; Pyne, S.G. Antimalarial and cytotoxic activities of pregnene-type steroidal alkaloids from Holarrhena pubescens roots. Nat. Prod. Res. 2017, 33, 782–788. [Google Scholar] [CrossRef]
  19. Zhou, L.N.; Ge, X.L.; Dong, T.T.; Gao, H.; Sun, B.H. Antibacterial steroidal alkaloids from Holarrhena antidysenteriaca. Chin. J. Nat. Med. 2017, 15, 540–545. [Google Scholar] [CrossRef]
  20. Tschesche, R.; Ockenfels, H. Uber, kurchi-alkaloide, V. 7α-hydroxy-conessin und holonamin, zwei neue basen aus kurchi-rinde. Chem. Ber. 1964, 97, 2316–2325. [Google Scholar] [CrossRef]
  21. Bhutani, K.K.; Vaid, R.M.; Ali, M.; Kapoor, R.; Soodan, S.R.; Kumar, D. Steroidal alkaloids from Holarrhena antidysenterica. Phytochemistry 1990, 29, 969–972. [Google Scholar] [CrossRef]
  22. Khalid, A.; Anjum, S.; Khan, M.R.; Choudhary, M.I. Kinetics and structure–activity relationship studies on pregnane-type steroidal alkaloids that inhibit cholinesterases. Bioorg. Med. Chem. 2004, 12, 1995–2003. [Google Scholar] [CrossRef]
  23. Kumari, G.K.; Rao, L.J.M.; Rao, K.R.; Rao, N.P.; Kaneko, K.; Mitsuhashi, H. Solanopubamides A and B, two further steroidal alkaloids from Solanum pubescens. Phytochemistry 2003, 25, 2003–2004. [Google Scholar] [CrossRef]
  24. Cheenpracha, S.; Jitonnom, J.; Komek, M.; Ritthiwigrom, T.; Laphookhieo, S. Acetylcholinesterase inhibitory activity and molecular docking study of steroidal alkaloids from Holarrhena pubescens barks. Steroids 2016, 108, 92–98. [Google Scholar] [CrossRef]
  25. Atta-Ur-Rahman; Choudhary, M.I.; Khan, M.R.; Anjum, S.; Farooq, A.; Iqbal, M.Z. New steroidal alkaloids from Sarcococca saligna. Nat. Prod. Res. 2003, 17, 235–241. [Google Scholar] [CrossRef]
  26. Gelaw, H. Isolation of crotepoxide from berries of Croton macrostachyus and evaluation of its anti-leishmanial activity. J. Pharmacogn. Phytochem. 2012, 1, 15–24. [Google Scholar]
  27. Srisurichan, S.; Pornpakakul, S. Triterpenoids from the seedpods of Holarrhena curtisii King and Gamble. Phytochem. Lett. 2015, 12, 282–286. [Google Scholar] [CrossRef]
  28. Zeb, M.A. Isolation and biological activity of β-Sitosterol and Stigmasterol from the roots of Indigofera heterantha. Pharm. Pharmacol. Intern. J. 2017, 5, 204–207. [Google Scholar] [CrossRef]
  29. Choudhary, M.I.; Ali, A.; Wahab, A.; Khan, S.; Rasheed, S.L.; Shyaula, R.A. New antiglycation and enzyme inhibitors from Parmotrema cooperi. Sci. China Chem. 2011, 54, 1926–1931. [Google Scholar] [CrossRef]
  30. Hong, Y.H.; Wang, S.C.; Hsu, C.; Lin, B.F.; Kuo, Y.H.; Huang, C.J. Phytoestrogenic compounds in alfalfa sprout (Medicago sativa) beyond coumestrol. J. Agric. Food Chem. 2011, 59, 131–137. [Google Scholar] [CrossRef] [PubMed]
  31. Díaz, A.B.; Vera, J.; Cote, V.; Bruno-Colmenárez, I.; De Delgado, G.D. NMR elucidation and crystal structure analysis of 1-hydroxy-3,6-dimethoxy-8-methyl-9H-xanthen-9-one (lichexanthone) isolated from Vismia baccifera (Guttiferae). Bol. Latinoam. Caribe Plantas Med. Aromat. 2010, 9, 470–474. [Google Scholar]
  32. Egbubine, C.O.; Adeyemi, M.M.; Habila, J.D. Isolation and characterization of betulinic acid from the stem bark of Feretia canthioides Hiern and its antimalarial potential. Bull. Natl. Res. Cent. 2020, 44, 441–447. [Google Scholar] [CrossRef]
  33. Lunardi, I.; Peixoto, J.L.B.; Silva, C.C.D.; Shuquel, I.T.A.; Basso, E.A.; Vidotti, G.J. Triterpenic acids from Eugenia moraviana. J. Braz. Chem. Soc. 2001, 12, 180–183. [Google Scholar] [CrossRef]
  34. Castellano, J.M.; Ramos-Romero, S.; Perona, J.S. Oleanolic acid: Extraction, characterization and biological activity. Nutrients 2022, 14, 623. [Google Scholar] [CrossRef]
  35. Birhanu, G. Isolation of ursolic acid from the leaves of Ocimum lamiifolium collected from Addis Ababa Area, Ethiopia. Afr. J. Biotechnol. 2020, 19, 65–70. [Google Scholar] [CrossRef]
  36. Xing-Gen, T.; Xiao-Rong, Z.; Shu-Lin, P.; Xun, L.; Li-Sheng, D. Chemical constituents from the roots of Biondia hemsleyana. Chem. J. Chin. Univ. 2003, 24, 436–441. [Google Scholar]
  37. Baniadam, S.; Rahiminejad, M.R.; Ghannadian, M.; Saeidi, H.; Ayatollahi, A.; Aghaei, M.M. Cycloartane triterpenoids from Euphorbia Macrostegia with their cytotoxicity against MDA-MB48 and MCF-7 cancer cell lines. Iran. J. Pharm. Res. IJPR 2014, 13, 135–141. [Google Scholar]
  38. Prabowo, W.C.; Wirasutisna, K.R.; Insanu, M. Isolation and characterization of 3-acetyl aleuritolic acid and scopoletin from stem bark of Aleurites moluccana (L.) Willd. Int. J. Pharm. Pharm. Sci. 2013, 5, 851–853. [Google Scholar]
  39. Singh, N.; Kaushik, N.K.; Mohanakrishnan, D.; Tiwari, S.K.; Sahal, D. Antiplasmodial activity of medicinal plants from Chhotanagpur plateau, Jharkhand, India. J. Ethnopharmacol. 2015, 13, 152–162. [Google Scholar] [CrossRef]
  40. Sajjadi, S.E.; Ghanadian, M.; Haghighi, M.; Mouhebat, L. Cytotoxic effect of Cousinia verbascifolia Bunge against OVCAR-3 and HT-29 cancer cells. J. HerbMed Pharmacol. 2015, 4, 15–19. [Google Scholar]
  41. Dua, V.K.; Verma, G.; Singh, B.; Rajan, A.; Bagai, U.; Agarwal, D.D.; Gupta, N.; Kumar, S.; Rastogi, A. Anti-malarial property of steroidal alkaloid conessine isolated from the bark of Holarrhena antidysenterica. Malar. J. 2013, 12, 194. [Google Scholar] [CrossRef]
  42. Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef] [PubMed]
  43. Elhag, I.Y. Chapter 12 Role of AI in ADME/Tox toward formulation optimization and delivery. In A Handbook of Artificial Intelligence in Drug Delivery; Academic Press: Cambridge, MA, USA, 2023; pp. 301–345. [Google Scholar] [CrossRef]
  44. Banerjee, P.; Kemmler, E.; Dunkel, M.; Preissner, R. ProTox 3.0: A webserver for the prediction of toxicity of chemicals. Nucleic Acids Res. 2024, 52, W513–W520. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Base Peak Chromatogram (BPC) of the alkaloid fraction from the stem bark of H. floribunda.
Figure 1. Base Peak Chromatogram (BPC) of the alkaloid fraction from the stem bark of H. floribunda.
Biomolecules 15 01415 g001
Figure 2. SNAP-MS networking of the alkaloid fraction of the stem bark of H. floribunda.
Figure 2. SNAP-MS networking of the alkaloid fraction of the stem bark of H. floribunda.
Biomolecules 15 01415 g002
Figure 3. Structures of steroidal alkaloids identified from the stem bark of H. floribunda (117).
Figure 3. Structures of steroidal alkaloids identified from the stem bark of H. floribunda (117).
Biomolecules 15 01415 g003
Figure 4. Structures of isolated compounds from the stem bark of H. floribunda.
Figure 4. Structures of isolated compounds from the stem bark of H. floribunda.
Biomolecules 15 01415 g004
Figure 5. Pie chart of target prediction of compounds (5, 14, and 16).
Figure 5. Pie chart of target prediction of compounds (5, 14, and 16).
Biomolecules 15 01415 g005
Table 1. Compounds identified from the stem bark of H. floribunda.
Table 1. Compounds identified from the stem bark of H. floribunda.
RT (min)[M + H]+
Exp
[M + H]+
Theo
MS/MS PatternsNamesReferences
12.6343.3113343.3108312, 269Isoconessimine (1)[17]
24.7359.3059359.3057341, 191Regholarrhenine D (2)[18]
37.0313.2684313.2638/Irheline (3)[18]
47.5329.2949329.2951269Conimin/conamine (4)[19]
59.3357.3275357.3264312, 269Conessine (5)[10]
612.5343.3163343.3108298, 269Conessimine (6)[17]
712.8373.3259373.3214312, 2697α-hydroxyconessine (7)[20]
814.0405.3402405.3476312, 267Regholarrhenine E (8)[21]
915.0441.3476441.3476327, 267, 157Salignemamide D (9)[22]
1015.2469.3793469.3789455, 424, 343Holarrhetine (10) [17]
1115.3455.3653455.3632267, 341Holarrhesine (11)[17]
1215.5457.3781457.3789441, 427Solanopubamide B (12)[23]
1316.0469.3778469.3789455, 441, 424Holarrhetine isomer (13)/
1418.7316.2674316.2635161Holaphyllamine (14)[24]
1519.2318.2796318.2791161, 261Holaphyllaminol (15)[24]
1619.3330.2789330.2791261, 161, 316N-methylholaphyllamine (16)[10]
1719.4441.3476441.3476327, 267Salignemamide D isomer (17)[25]
Salignemamide D (9) and Solanopubamide B (12) are reported here for the first time from the genus Holarrhena. However, they were previously isolated from Solanum pubescens (Solanaceae) and Sarcococca saligna (Buxaceae), respectively [21,25].
Table 2. In Vitro antiplasmodial and cytotoxicity results of extract, fractions, and compounds from the stem bark of H. floribunda.
Table 2. In Vitro antiplasmodial and cytotoxicity results of extract, fractions, and compounds from the stem bark of H. floribunda.
Extract/Fractions/
Compounds
IC50/Pf3D7IC50)/PfDd2RI: [IC50PfDd2/IC50Pf3D7]CC50 on Vero CellsCC50 on KB-3-1
IC50 Values [Compound and Reference Drug (µM) and Extracts and Fractions (µg/mL)]
EtOH crude extract14.11 ± 3.0623.32 ± 2.391.65>500 µg/mLND
n-hexane fraction36.36 ± 1.5943.54 ± 2.74 µg/mL1.19>500 µg/mLND
CH2Cl2 fraction>100>100->500 µg/mLND
n-butanol fractionNTNT-NDND
Alkaloid fraction6.54 ± 0.5708.27 ± 0.6501.26162.9 ± 3.970 µg/mLNT
conessine (5)25.97 ± 0.4328.51 ± 0.7041.09NTNT
Irheline (3)>50>50-NTNT
Holarrhesine (11)NTNT-NTNT
Holaphyllamine (14)49.53 ± 4.83055.78 ± 9.750 µM1.12NT9.8 µM
Holaphylaminol (15)NTNT-NTNT
Betulinic acid (22)NTNT-NTNT
Ethylorsalinate (26)>50>50-NTNT
Lichexanthone (27)33.41 ± 1.20036.46 ± 1.391.09NTNT
Chloroquine *0.045 ± 0.0030.73 ± 0.09016.22NTNT
Artemisinin *0.035 ± 0.0000.025 ± 0.0060.71NTNT
Cryptophycin *----1.3 × 10−5 μM
IC50 = half inhibitory concentration, CC50 = cytotoxic concentration 50%, * = Reference drug, NT = Not Tested, RI = Resistance Index.
Table 3. In vitro antiplasmodial activity of some identified and isolated steroidal alkaloids reported in the literature.
Table 3. In vitro antiplasmodial activity of some identified and isolated steroidal alkaloids reported in the literature.
NamesIC50 (µM)References
Isoconessimine (1)3.39 ± 079 (Pf FCB1)[17]
Irheline (3)1.2 (Pf K1)[18]
Conimin (4)8.0 (Pf K1)
Conessine (5)1.04 ± 014 (Pf FCB1)[17]
Holarrhetine (10)1.13 ± 0.32 (Pf FCB1)
Holarrhesine (11)0.97 ± 0.11 (Pf FCB1)
N-methylholaphyllamine (16)10.6 (Pf K1)[18]
Holaphyllaminol (15)11.7 (Pf K1)
Chloroquine *0.13 ± 0.03[17]
IC50 = half inhibitory concentration, Pf: Plasmodium falciparum, * = Reference drug.
Table 4. Drug-likeness, physicochemical parameters and lead-likeness of some alkaloids identified from the root and the stem bark of H. floribunda.
Table 4. Drug-likeness, physicochemical parameters and lead-likeness of some alkaloids identified from the root and the stem bark of H. floribunda.
Holarrhesine (11)Irheline (3)Holarrhetine (10)Holaphyllaminol (15)Holaphyllamine (14)Conessine (5)N-methylholaphyllamine (16)Isoconessimine (1)Conimin (4)
Physicochemical Property
MW (g/mol)454.69312.49468.71317.51315.49356.59329.52342.56328.53
Pharmacokinetics
GI absorptionHighHighHighHighHighHighHighHighHigh
BBB permeantYesYesYesYesNoYesYesYesYes
P-gp substrateNoNoNoYesNoNoNoNoNo
CYP1A2 inhibitorNoNoNoNoNoNoNoNoNo
CYP2C19 inhibitorNoNoNoNoNoNoNoNoNo
CYP2C9 inhibitorNoNoNoNoNoNoNoNoNo
CYP2D6 inhibitorNoNoNoNoNoNoNoNoNo
CYP3A4 inhibitorNoNoNoNoNoNoNoNoNo
Lipophilicity
Log Kp (skin permeation)−5.24 cm/s−6.15 cm/s−4.99 cm/s−5.20 cm/s−5.41 cm/s−5.03 cm/s−5.14 cm/s−5.27 cm/s−5.52 cm/s
Log S (ESOL)−5.73−3.60−6.11−4.44−4.22−5.04−4.57−4.66−4.28
Solubility8.45 × 10−4 mg/mL; 1.86 × 10−6 mol/L7.88 × 10−2 mg/mL; 2.52 × 10−4 mol/L3.60 × 10−4 mg/mL; 7.69 × 10−7 mol/L1.16 × 10−2 mg/mL; 3.64 × 10−5 mol/L1.88 × 10−2 mg/mL; 5.96 × 10−5 mol/L3.25 × 10−3 mg/mL; 9.11 × 10−6 mol/L8.93 × 10−3 mg/mL; 2.71 × 10−5 mol/L7.43 × 10−3 mg/mL; 2.17 × 10−5 mol/L1.72 × 10−2 mg/mL; 5.24 × 10−5 mol/L
ClassModerately solubleSolublePoorly solubleModerately solubleModerately solubleModerately solubleModerately solubleModerately solubleModerately soluble
Log S (Ali)−6.03−3.36−6.33−4.96−3.90−4.72−4.80−4.43−4.12
Solubility4.27 × 10−4 mg/mL; 9.38 × 10−7 mol/L1.38 × 10−1 mg/mL; 4.40 × 10−4 mol/L2.19 × 10−4 mg/mL; 4.67 × 10−7 mol/L3.45 × 10−3 mg/mL; 1.09 × 10−5 mol/L3.94 × 10−2 mg/mL; 1.25 × 10−4 mol/L6.79 × 10−3 mg/mL; 1.90 × 10−5 mol/L5.21 × 10−3 mg/mL; 1.58 × 10−5 mol/L1.38 × 10−2 mg/mL; 4.04 × 10−5 mol/L2.47 × 10−2 mg/mL; 7.51 × 10−5 mol/L
Drug-likeness
LipinskiYes; 1 violation: MLOGP > 4.15Yes; 0 violationYes; 1 violation: MLOGP > 4.15Yes; 1 violation: MLOGP > 4.15Yes, 0 violationYes; 1 violation: MLOGP > 4.15Yes; 1 violation: MLOGP > 4.15Yes; 1 violation: MLOGP > 4.15Yes; 1 violation: MLOGP > 4.15
GhoseNo; 2 violations: MR > 130, atoms > 70YesNo; 2 violations: MR > 130, atoms > 70YesYesYesYesYesYes
VeberYesYesYesYesYesYesYesYesYes
EganYesYesYesYesYesYesYesYesYes
MueggeNo; 1 violation: XLOGP3 > 5YesNo; 1 violation: XLOGP3 > 5YesYesYesYesYesYes
Medicinal Chemistry
Bioavailability Score0.550.550.550.550.550.550.550.550.55
Lead-likenessNo; 2 violations: MW > 350, XLOGP3 > 3.5YesNo; 2 violations: MW > 350, XLOGP3 > 3.5No; 1 violation: XLOGP3 > 3.5No; 1 violation: XLOGP3 > 3.5No; 2 violations: MW > 350, XLOGP3 > 3.5No; 1 violation: XLOGP3 > 3.5No; 1 violation: XLOGP3 > 3.5No; 1 violation: XLOGP3 > 3.5
MW: molecular weight, Log s: aqueous solubility (scale: <−10 < Poorly < −6 < Moderately <−4 < soluble < −2 very < 0<Highly); BBB: Blood–brain barrier; Lipinsky (Criteria: MW ≤ 500; MLOGP ≤ 4.15; N or O ≤ 10; NH or OH ≤5); Ghose (Criteria: 160 ≤ MW ≤ 480; −0.4 ≤ WLOGP ≤ 5.6; 40 ≤ MR ≤ 130; 20 ≤ atoms ≤ 70); Veber (criteria: rotatable bonds ≤ 10, TPSA ≤ 140); Egan (criteria: WLOGP ≤ 5.88, TPSA ≤ 131.6); Muegge, Num. ring ≤ 7, Num. carbon > 4, Num. heteroatom > 1, num. rotatable bonds ≤ 15, H-bond acc ≤ 10, H-bond don. ≤ 5200 ≤ MW ≤ 600, −2 ≤ XLOGP ≤ 5); Lead-likeness: 250 ≤ MW ≤ 350, XLOGP ≤ 3.5, Num. rotatable bonds ≤ 7), GI absorption: gastrointestinal absorption.
Table 5. In Silico acute toxicity of bioactive compounds.
Table 5. In Silico acute toxicity of bioactive compounds.
Targets CompoundsHepatotoxicity (SP)Nephrotoxicity (SP)Cardiotoxicity (SP)Respiratory Toxicity (SP)LD50
(mg/kg)
Conesine (5)Inactive (0.89)Inactive (0.94)Inactive (0.81)Active (0.77)750
Holaphyllamine (14)Inactive (0.75)Inactive (0.92)Inactive (0.78)Active (0.73)390
Lichexanthone (27)Inactive (0.75)active (0.61)Active (0.53)Active (0.75)3200
SP: Score of the prediction; Average for active compounds: Hepatotoxicity: 0.82; Nephrotoxicity: 0.75; Cardiotoxicity: 0.86; Respiratory toxicity: 0.78; Class I: fatal if swallowed (LD50 ≤ 5 mg/kg); Class II: fatal if swallowed (5 mg/kg < LD50 ≤ 50 mg/kg); Class III: toxic if swallowed (50 mg/kg < LD50 ≤ 300 mg/kg); Class IV: harmful if swallowed (300 mg/kg < LD50 ≤ 2000 mg/kg); Class V: may be harmful if swallowed (2000 mg/kg < LD50 ≤ 5000 mg/kg); Class VI: non-toxic (LD50 > 5000 mg/kg).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Djila Possi, F.L.; Kinyok, M.J.; Mbasso Tameko, J.E.; G. Mountessou, B.Y.; Jumeta Dongmo, J.K.; Tchatat Tali, M.B.; Kene Dongmo, A.; Fekam Boyom, F.; Kezetas Bankeu, J.J.; Sewald, N.; et al. UHPLC-QTOF-ESI-MS/MS, SNAP-MS Identification, In Silico Prediction of Pharmacokinetic Properties of Constituents from the Stem Bark of Holarrhena floribunda (G. Don) T. Durand and Schinz (Apocynaceae). Biomolecules 2025, 15, 1415. https://doi.org/10.3390/biom15101415

AMA Style

Djila Possi FL, Kinyok MJ, Mbasso Tameko JE, G. Mountessou BY, Jumeta Dongmo JK, Tchatat Tali MB, Kene Dongmo A, Fekam Boyom F, Kezetas Bankeu JJ, Sewald N, et al. UHPLC-QTOF-ESI-MS/MS, SNAP-MS Identification, In Silico Prediction of Pharmacokinetic Properties of Constituents from the Stem Bark of Holarrhena floribunda (G. Don) T. Durand and Schinz (Apocynaceae). Biomolecules. 2025; 15(10):1415. https://doi.org/10.3390/biom15101415

Chicago/Turabian Style

Djila Possi, Franck Landry, Mc Jesus Kinyok, Joseph Eric Mbasso Tameko, Bel Youssouf G. Mountessou, Johanne Kevine Jumeta Dongmo, Mariscal Brice Tchatat Tali, Appolinaire Kene Dongmo, Fabrice Fekam Boyom, Jean Jules Kezetas Bankeu, Norbert Sewald, and et al. 2025. "UHPLC-QTOF-ESI-MS/MS, SNAP-MS Identification, In Silico Prediction of Pharmacokinetic Properties of Constituents from the Stem Bark of Holarrhena floribunda (G. Don) T. Durand and Schinz (Apocynaceae)" Biomolecules 15, no. 10: 1415. https://doi.org/10.3390/biom15101415

APA Style

Djila Possi, F. L., Kinyok, M. J., Mbasso Tameko, J. E., G. Mountessou, B. Y., Jumeta Dongmo, J. K., Tchatat Tali, M. B., Kene Dongmo, A., Fekam Boyom, F., Kezetas Bankeu, J. J., Sewald, N., Chouna, J. R., & Ndjakou Lenta, B. (2025). UHPLC-QTOF-ESI-MS/MS, SNAP-MS Identification, In Silico Prediction of Pharmacokinetic Properties of Constituents from the Stem Bark of Holarrhena floribunda (G. Don) T. Durand and Schinz (Apocynaceae). Biomolecules, 15(10), 1415. https://doi.org/10.3390/biom15101415

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

Article Metrics

Back to TopTop