Skip to main content

In vivo antimalarial effect of 1-hydroxy-5,6,7-trimethoxyxanthone isolated from Mammea siamensis T. Anders. flowers: pharmacokinetic and acute toxicity studies

Abstract

Background

The potent antiplasmodial activity of 1-hydroxy-5,6,7-trimethoxyxanthone (HTX), isolated from Mammea siamensis T. Anders. flowers, has previously been demonstrated in vitro. However, its in vivo activity has not been reported. Therefore, this study aimed to investigate the antimalarial activity and acute toxicity of HTX in a mouse model and to evaluate the pharmacokinetic profile of HTX following a single intraperitoneal administration.

Methods

The in vivo antimalarial activity of HTX was evaluated using a 4-day suppressive test. Mice were intraperitoneally injected with Plasmodium berghei ANKA strain and given HTX daily for 4 days. To detect acute toxicity, mice received a single dose of HTX and were observed for 14 days. Additionally, the biochemical parameters of the liver and kidney functions as well as the histopathology of liver and kidney tissues were examined. HTX pharmacokinetics after intraperitoneal administration was also investigated in a mouse model. Liquid chromatography triple quadrupole mass spectrometry was used to quantify plasma HTX and calculate pharmacokinetic parameters with the PKSolver software.

Results

HTX at 10 mg/kg body weight significantly suppressed parasitemia in malaria-infected mice by 74.26%. Mice treated with 3 mg/kg HTX showed 46.88% suppression, whereas mice treated with 1 mg/kg displayed 34.56% suppression. Additionally, no symptoms of acute toxicity were observed in the HTX-treated groups. There were no significant alterations in the biochemical parameters of the liver and kidney functions and no histological changes in liver or kidney tissues. Following intraperitoneal HTX administration, the pharmacokinetic profile exhibited a maximum concentration (Cmax) of 94.02 ng/mL, time to attain Cmax (Tmax) of 0.5 h, mean resident time of 14.80 h, and elimination half-life of 13.88 h.

Conclusions

HTX has in vivo antimalarial properties against P. berghei infection. Acute toxicity studies of HTX did not show behavioral changes or mortality. The median lethal dose was greater than 50 mg/kg body weight. Pharmacokinetic studies showed that HTX has a long elimination half-life; hence, shortening the duration of malaria treatment may be required to minimize toxicity.

Peer Review reports

Background

Malaria is a major cause of morbidity and mortality throughout tropical and subtropical regions [1]. The disease is caused by protozoans from the genus Plasmodium. The most virulent parasite species responsible for severe clinical malaria and death is Plasmodium falciparum [2]. According to a World Health Organization report, in 2022, there were an estimated 247 million malaria cases and 625,000 deaths worldwide by 2020 [3]. Although artemisinin combined with other drugs, known as artemisinin-based combination therapies (ACTs), is the most effective way to treat malaria, reduced sensitivity, as evidenced by the slow clearance of P. falciparum parasites, has been reported. Artemisinin resistance was first reported in Cambodia in 2006 and has since spread to most of Southeast Asia. ACTs failures are increasing in the Greater Mekong Region, reaching 50%, which was observed in 140 patients when dihydroartemisinin-piperaquine was recently tested in 2015–2018 [4, 5]. Plasmodium resistance, especially P. falciparum, to currently available drugs poses a significant problem for treating and preventing malaria [6]. Increased resistance of malaria parasites to available drugs has encouraged researchers to continue searching for new and more effective antimalarials [7].

Medicinal plants are a potential source of new antimalarial drugs as they contain numerous compounds with diverse structures and pharmacological activities. These metabolites can be optimized to obtain better therapeutic agents [8]. Furthermore, medicinal plants have been used in traditional malaria treatment because of their efficacy, safety, low cost, and availability [9, 10]. Therefore, there is a need to identify new chemical compounds with antimalarial activity, especially from natural products, including plants and microorganisms, such as bacteria and fungi, which are rich sources of many bioactive substances [11]. The first antimalarial drug identified was quinine, which was originally isolated from the bark of the Cinchona species [12]. The current antimalarial drug of choice is artemisinin (Qinghaosu), which was initially derived from the leaves of Artemisia annua L [13].

Mammea siamensis T. Anders, locally known as Saraphi in Thailand, belongs to the Calophyllaceae family. Its flowers are traditionally used for treating heart problems and fever as well as to increase appetite [14]. A methanolic extract of the flowers of M. siamensis and its constituents have been reported to possess aromatase inhibitory activity [15], antiproliferative and apoptotic effects [16, 17], and the ability to suppress nitric oxide (NO) production [18]. In addition, our previous study found that the methanolic extract of the flowers of this plant exhibits antimalarial activity against the Plasmodium falciparum K1 strain with a half-maximum inhibitory concentration (IC50) value of 1.50 mg/mL [19]. In an in vitro study, 1-hydroxy-5,6,7-trimethoxyxanthone (HTX), its isolated compound, demonstrated strong antiplasmodial activity with an IC50 value of 9.57 µM [20]. This compound has not been tested for its efficacy in an in vivo model. Therefore, the present study aimed to investigate the antimalarial activity and acute toxicity of HTX in a mouse model and to evaluate the pharmacokinetic profile of this compound.

Methods

Plant materials

M. siamensis flowers were purchased from a Thai traditional drug store in Nakhon Si Thammarat Province, Thailand. The collection of plant materials followed the relevant guidelines and regulations of The Plant Varieties Protection, Department of Agriculture, Ministry of Agriculture and Cooperatives, Thailand. The plant was identified by Assoc. Prof. Tanomjit Supavita, a botanist at the School of Pharmacy at Walailak University. The plant material was deposited at the School of Medicine, Walailak University, with identification voucher number SMD122006002. Plant materials were rinsed with distilled water before being dried in a hot air oven at 60 °C and pulverized to coarse powder using a herb grinder.

General experimental procedure

Silica gel (230–400 mesh, Sili Cycle Inc., Canada) was used for column chromatography (CC). All solvents were purchased from Labscan (Thailand) and were of analytical grade. All reagents were purchased from Sigma-Aldrich (USA). The nuclear magnetic resonance (NMR) spectra were obtained using an Avance NEO spectrometer (Bruker) at 500 MHz for 1H.

Extraction and isolation of compounds

The dried M. siamensis flower powder (8.0 kg) was macerated extensively in dichloromethane (15 L) for 72 h with occasional shaking. The solvent was filtered through Whatman No. 1 filter paper. The residue was re-macerated for 72 h, and this procedure was repeated thrice. The filtrates were combined and concentrated using a rotary evaporator (Rotavapor®, Buchi, China) at 50 °C to obtain a brownish viscous crude extract (76.8 g). Finally, the dried crude extract was kept in screw-cap containers and stored at 4 °C for further use.

Based on its in vitro antiplasmodial activity, HTX was isolated according to our previous study, with some modifications [20]. The dichloromethane extract (75 g) underwent quick CC (QCC) through silica gel by step-gradient elution starting with dichloromethane. The polarity was increased with ethyl acetate (EtOAc) and acetone to yield 12 fractions (F1-F12), based on thin-layer chromatography profiles. Fraction 6 (8.2 g) was purified by QCC using 10% EtOAc in hexane to produce five subfractions (6A-6E). Subfraction 6B (4.13 g) was further purified by QCC using 15% EtOAc in hexane to obtain four subfractions (6B1-6B4). Subfraction 6B2 (2.66 g) was further purified by CC with 15% EtOAc in hexane to obtain compound 1 as a yellow powder (62.6 mg).

Animals

Healthy adult male Institute of Cancer Research (ICR) mice weighing 25–30 g body weight and aged 6–8 weeks were obtained from Nomura Siam International Co., Ltd., Pathumwan, Bangkok, Thailand, for antimalarial activity, acute toxicity, and pharmacokinetic testing. The study was approved by the Animal Ethics Committee of Walailak University (certificate number: WU-ACUC-65015), and in accordance with Ethical Principles and Guidelines for the Use of Animals by the National Research Council of Thailand. All mice were housed in cages under standard environmental conditions at a relative humidity of 50–60% and a room temperature of about 22 °C (± 3 °C) under a natural 12-hour dark-light cycle. Mice had free access to pellet diet and clean drinking water. All mice were acclimatized to the experimental environment for a week before beginning the experiment. This study was reported in accordance with ARRIVE guidelines.

Parasite inoculation

Wild-type P. berghei ANKA strain was obtained from Thomas F. McCutchan (BEI Resources, NIAID, NIH). P. berghei-infected red blood cells were administered intraperitoneally to donor mice. When parasitemia levels reached 20–30%, blood was collected from the heart using a cardiac puncture procedure and placed in a vacutainer heparinized tube for injection into experimental animals [21].

In vivo 4-day suppressive test of P. berghei

A 4-day suppressive test was used to measure the schizonticidal activity of HTX in P. berghei-infected ICR mice. This test was performed according to a previous method, with some modifications [22, 23]. Male ICR mice were randomly divided into six groups, each consisting of five mice, as shown in Table 1. All mice were intraperitoneally injected with 1 × 107P. berghei ANKA-parasitized erythrocytes. Treatment was initiated four hours after the mice were infected with Plasmodium parasite on the first day (D0). The negative control group was administered 150 µl of 2% dimethyl sulfoxide (DMSO) in normal saline solution (NSS), whereas the positive control group was administered 3 mg/kg body weight of artesunate and chloroquine intraperitoneally each day. For each experimental group, animals received daily intraperitoneal doses of 1, 3, or 10 mg/kg body weight of HTX. Mice received each substance daily for 4 days (D0-D3). On the fifth day (D4), parasitemia was determined using Giemsa-stained thin blood smears prepared from the tails of each mouse. Mice were euthanized at the end of the experiment by intraperitoneal injection of pentobarbital (200 mg/kg body weight).

Finally, the percentage suppression of parasite growth was calculated using the following equation:

$$\% {\text{inhibition = }}\frac{\begin{gathered}{\text{(parasitemia of negative group - }} \hfill \\{\text{ parasitemia of treated group)}} \hfill \\ \end{gathered} }{{{\text{(parasitemia of negative group)}}}}{\text{ }} \times {\text{ 100}}$$
Table 1 Group classifications and doses used in the 4-day suppressive test

Acute toxicity test

Acute HTX toxicity was determined in non-infected ICR mice aged 6–8 weeks and weighing 25–35 g using the Organization for Economic Cooperation and Development standard criteria [24]. Fifteen mice were randomly divided into three groups (n = 5): untreated, negative control, and HTX-treated. All mice fasted for three hours prior to the experiment, and only drinking water was available. HTX was dissolved in 2% DMSO in NSS at 50 mg/kg body weight. In the treatment group, the first mouse was intraperitoneally given a single dose of 50 mg/kg body weight HTX, whereas in the negative control group, the first mouse was intraperitoneally given 200 µL of 2% DMSO in NSS. After dosing, mice were observed for 24 h for gross physiological and behavioral changes, such as sleep, hair erection, poor appetite, convulsions, diarrhea, lacrimation, salivation, mortality, and other manifestations of toxicity. If death was not observed within 24 h in the first mouse, the same dose was administered to 4 more mice, and they were observed for signs and symptoms of toxicity for 14 days. All mice were anaesthetized using pentobarbital (60 mg/kg body weight) by intraperitoneal injection. Blood samples were collected via cardiac puncture into heparinized tubes for biochemical analyses. The liver and kidney tissues were collected for histological examination. The mice were subsequently euthanized with an intraperitoneal injection of pentobarbital (200 mg/kg body weight).

Histopathological examination

Histopathological examinations of liver and kidney tissues were performed in accordance with standard histological protocols, as reported previously [25, 26]. In brief, all tissues were fixed in 10% buffered formalin, dehydrated using a gradient series of ethanol solutions, cleaned with xylene, and deposited in a paraffin mold. The paraffin blocks were then sectioned to a thickness of 5 μm using a rotary microtome, transferred to glass slides, and stained with hematoxylin and eosin (H&E) solution. The stained slides were examined under light microscopy by two independent observers who were blinded to the experimental groups for examining the histological changes.

In vivo pharmacokinetic study

Samples preparation

A pharmacokinetic study was conducted in non-infected ICR mice aged 6–8 weeks and weighing 25–35 g. Male ICR mice were randomly divided into two groups, each consisting of five mice. The first group is a negative control group and was injected with 150 µL of 2% DMSO in NSS, whereas the second group is a test group that was administered 5 mg/kg body weight of HTX compound intraperitoneally. Blood (50 L) was taken from the tail vein prior to administration of test compounds (0 h) and after at, 0.5, 1, 2, 3, 5, 8, 24, 48, and 72 h. Blood samples were collected in microtubes containing ethylenediaminetetraacetic acid (EDTA) and kept on ice until they were centrifuged at 3,000 × g for 10 min at 4 °C to allow the collection of plasma, which was stored at − 80 °C.

Liquid chromatography triple quadrupole mass spectrometry (LC-MS/MS)

LC-MS/MS (Agilent 1290 infinity LC and Agilent 6490 triple quadrupole mass spectrometer) with an electrospray ionization (ESI, Agilent Technologies, USA) source was used to detect HTX in the plasma. Using a VertiSepTM USP C18 column (4.6 mm × 150 mm, particle size = 5 μm; Vertical Chromatography Co., ltd., Nonthaburi, Thailand), the separation was achieved. Acetonitrile:1 mM formic acid (7:3) was used as the mobile phase and was maintained at a constant flow rate of 0.5 mL/min. The injection volume was 1 µL, and column temperature was maintained at 30 °C. The following operating conditions for the MS were optimized: the source temperature was kept at 200 °C, the ion spray voltage for the positive mode was set at 3 KV, and the collision energy for HTX was set at 43 V. Nitrogen was used as the collision gas. The nebulizer and sheath gas flow rates were set to 14 and 11 L/min, respectively. The MassHunter qualitative analysis software was used to acquire the data (Agilent Technologies, Inc. Headquarters, CA, USA). HTX was quantified using the transitions for HTX at m/z 303→199 in the multiple reaction monitoring (MRM) mode.

Preparation of the standard and quality control samples

By dissolving precisely weighed HTX and reference compounds in acetonitrile, stock HTX solutions were prepared. By diluting the stock solutions using a mixture of acetonitrile and water (2:8, v/v), a series of working solutions with concentrations between 7.81 and 1000 ng/mL were prepared. All solutions were stored at 4 °C. By spiking the blank plasma (50 µL) with 25 µL of standard working solutions, HTX calibration standards (0.78, 1.56, 3.13, 6.25, 12.5, 25.0, 50, 125, and 250 ng/mL) were prepared. The extraction solvent ethyl acetate (0.5 mL) was added to 75 µL of HTX-spiked plasma. The sample was vortexed for 1 min, followed by ultrasonic vibration for 15 min. Then, centrifugation for 10 min at 15,000 × g was done. The upper organic layer was transferred to a new microtube and dried via evaporation. The dried extract was reconstituted in 150 µL of 50% (v/v) acetonitrile in water. HTX recovery experiments were performed to confirm extraction efficacy. The effectiveness of liquid-liquid extraction was then determined by comparing the peak regions of the extracted HTX-spiked plasma with those of HTX spiked with blank plasma extract.

Pharmacokinetic analysis of HTX

The plasma concentration-time profiles of HTX were determined by non-compartmental analysis with PKSolver 2.0 software. Pharmacokinetic parameters were calculated to estimate the areas under the concentration-time curve (AUCs) from 0 to 48 h and from 0 to ∞, elimination half-life (t1/2), maximum concentration achieved (Cmax), time to attain Cmax (Tmax), mean residence time (MRT), apparent volume of distribution, and clearance. Additionally, the apparent volume of the central or plasma compartment, apparent volume of the peripheral compartment, transfer rate constant from the central compartment to the peripheral compartment, and transfer rate constant from the peripheral to central compartment were calculated using the two-compartment model.

Statistical analysis

Results were presented as means ± SEM. IBM SPSS Statistics version 23.0 software (SPSS, IL, USA) was used for statistical analysis. Normal distribution was tested using the Kolmogorov–Smirnov goodness-of-fit test. Statistical significance of parasitemia inhibition was determined using one-way ANOVA, followed by Tukey’s multiple comparison test. Statistical significance was set at 0.05 (p < 0.05).

Results

Identification of compound 1

The structures of compound 1 (Fig. 1) was elucidated by 1H-NMR analysis and confirmed by comparison with previously reported data [27]. Compound 1-hydroxy-5,6,7-trimethoxyxanthone (1): Yellow powder. 1H-NMR (acetone-d6, 500 MHz): δ 6.93 (1H, d, J = 8.3 Hz, H-2), 7.13 (1H, d, J = 8.3 Hz, H-4), 7.19 (1H, s, H-8), 7.68 (1H, t, J = 8.3 Hz, H-3), 12.68 (1H, brs, 1-OH), 3.94 (3H, s, 5-OCH3), 3.93 (3H, s, 6-OCH3), 3.95 (3H, s, 7-OCH3).

Fig. 1
figure 1

Structure of HTX compound

Antimalarial activity

The antimalarial effect of HTX was examined in a 4-day suppressive test as a standard model. Mice in each group received daily intraperitoneal doses of HTX at different concentrations (1, 3, and 10 mg/kg) body weight. The percentages of parasitemia and suppression are presented in Table 2. The results showed that HTX effectively suppressed Plasmodium parasites and showed significant (p < 0.05) antimalarial activity against P. berghei infection in mice compared to that of negative controls. The parasite counts decreased in a dose-dependent manner after HTX treatment. The highest level of suppression (74.26%) was obtained with a 10 mg/kg dose of HTX, followed by a dose of 3 mg/kg (46.88%), whereas the lowest level of suppression (34.56%) was obtained with a dose of 1 mg/kg. However, antimalarial activity against P. berghei infection at all doses was significantly (p < 0.05) lower than that of the positive control groups with standard drugs, artesunate and chloroquine (3 mg/kg each), which showed 91.26% and 92.07% suppression, respectively.

Table 2 Effects of HTX against Plasmodium parasites in malaria-infected mice

Acute toxicity

Acute toxicity study at 50 mg/kg HTX demonstrated no visible signs of physical and behavioral changes, such as sleep, hair erection, poor appetite, convulsions, diarrhea, lacrimation, salivation, mortality, and other manifestations of toxicity during the experimental period. No mortality was observed in any of the mice within 24 h of HTX administration or the following 14 days. Therefore, the lethal dose of HTX appears greater than 50 mg/kg body weight.

Effects of HTX on the liver and kidney functions

The effects of 50 mg/kg HTX on the biochemical parameters of the liver and kidneys are shown in Table 3. The biochemical parameters of liver function, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP), in the HTX-treated group were 46.20, 13.00, and 105.80 U/L. These were not significantly different from those in the untreated and negative controls (p > 0.05). Additionally, kidney function was evaluated based on plasma blood urea nitrogen (BUN) and creatinine levels, which were 16.00 and 0.25 mg/dL, respectively, in mice treated with 50 mg/kg HTX. These values were not significantly different compared to those of the untreated control (p > 0.05) after 14 days of exposure to HTX. These results showed that HTX did not affect the liver and kidney functions.

Table 3 Effects of HTX on the biochemical parameters of the liver and kidney functions in the acute toxicity test

Histopathological examination of the livers and kidneys of the mice

The liver and kidney sections from mice were analyzed histologically using H&E staining. Typical histological structures in different parts of the liver and kidneys of mice treated with 50 mg/kg HTX are shown in Fig. 2. The results demonstrated normal histology in the livers of mice treated with HTX, including the morphological characteristics of the hepatocytes, such as arrangement, size, and color staining. Moreover, inflammatory cell infiltration and sinusoidal vasodilation were not detected (Fig. 2E) when compared with the untreated control (Fig. 2A) and negative control groups (Fig. 2C). Histopathological changes in the kidneys were also investigated. The histological features of the kidneys in the HTX-treated group were also normal, including kidney epithelial cells, glomeruli, and Bowman’s capsules (Fig. 2F), compared with those in the untreated control (Fig. 2B) and negative control groups (Fig. 2D).

Fig. 2
figure 2

Histopathological examination of the liver and kidney sections of mice. Mice were intraperitoneally administered with 2% DMSO in normal saline solution (negative control) or HTX 50 mg/kg. Liver and kidney were excised and stained with hematoxylin and eosin. Untreated control group (A, B), negative control group (C, D), 50 mg/kg HTX-treated group (E, F). All images are 400 × magnification. Bars = 20 μm. T: tubules; G: glomerulus; CV: central vein; H: hepatocyte. Abbreviation: HTX, 1-hydroxy-5,6,7-trimethoxyxanthone

Mass spectrum

The mass spectrum of HTX [M + H]+ showed a peak at m/z 303 (Fig. 3A). The product ion with the highest relative abundance when the collision energy was optimized was at 198.8 m/z (Fig. 3B). The findings showed that the 43 V collision energy generated most of the aforementioned ion product, which was subsequently subjected to MRM mode analysis. HTX had a retention period of 4.126 min.

Fig. 3
figure 3

The molecular weight of HTX ([M-H]+, A) and the product ion of HTX (B). Abbreviation: HTX, 1-hydroxy-5,6,7-trimethoxyxanthone

Calibration, linearity, precision, and recovery

The calibration curve was linear for HTX concentrations in mouse plasma, ranging from 0.781 to 250 ng/mL, with a correlation value of R2 = 0.9978 (y = 1527x–1332). The precision of analysis in the concentration range was between 0.33 and 5.77% expressed as coefficient of variation. The signal-to-noise ratio at 0.781 ng/mL was 48.3 ± 8.1. Thus, a calibration curve was established with an HTX concentration range of 0.781–250 ng/mL. The LC-MS/MS system specifically detected HTX in plasma samples. The efficacy of the liquid-liquid extraction was confirmed (Table 4). The recovery was in the range of 87.2–106.3%; thus, the extraction method is efficient for recovering HTX from the sample.

Table 4 Recovery of HTX by liquid-liquid extraction in plasma

Pharmacokinetic analysis of HTX

Pharmacokinetic parameters of HTX are presented in Table 5. These results showed that HTX exhibited a Cmax of 94.02 ng/mL, Tmax of 0.5 h, MRT of 14.79 h, and a t1/2 of 13.88 h. The plasma concentration versus time profiles of HTX after intraperitoneal administration are illustrated in Fig. 4.

Table 5 Pharmacokinetic parameters of HTX
Fig. 4
figure 4

Plasma concentration-time curve of HTX following intraperitoneal administration at 5 mg/kg dose in ICR mice. Data expressed as mean ± standard deviation from five rats at different time points

Discussion

The present study investigated the antimalarial activity of HTX against P. berghei infection in mice as well as the acute toxicity and pharmacokinetic parameters. Here, the in vivo antimalarial activity of HTX was first investigated. Based on our previous study of the antimalarial properties of several compounds isolated from Mammea siamensis flowers, HTX was shown to exhibit the most potent antimalarial effect against the P. falciparum K1 strain. This is a chloroquine-resistant strain, with an IC50 value of 9.57 µM [20]. The antimalarial effects of the xanthone (HTX), showed a dose-dependent pattern. A high dose of HTX (10 mg/kg body weight) exhibited a 74.26% decrease in P. berghei.

Xanthones, secondary metabolites from natural sources, contain various substituents on two benzene rings, thus leading to their rich structural diversity. They possess a broad spectrum of pharmacological properties, such as anticancer, antitumor, antioxidant, antidiabetic, and anticarcinogenic activities [28,29,30,31,32]. Xanthones also exhibit antimalarial activity. Α-Mangostin, a xanthone from Garcinia mangostana, is active against the resistant P. falciparum chloroquine-resistant (FCR3) strain with an IC50 value of 0.2 µM [33]. A previous study reported that xanthones exhibit antimalarial effects by forming complexes with heme and inhibiting hemozoin formation [34]. Therefore, the antimalarial activity of HTX may have resulted from inhibition of hemozoin formation, which has the same mechanism of action as chloroquine, an antimalarial drug [35, 36].

Because in vivo toxicology data for HTX have never been reported, the acute intraperitoneal toxicity of HTX in ICR mice was investigated. This study was performed using 50 mg/kg HTX, and toxicity symptoms were monitored for 14 days. The results showed that until the end of the experiment, all treated mice were alive without any symptoms of hair erection, convulsions, diarrhea, lacrimation, and salivation or other manifestations of toxicity. Additionally, biochemical analyses were performed to assess the liver and kidney functions. Since the liver is the major target organ of toxicity, liver damage may compromise the integrity of hepatocytes, causing the release of membrane-bound enzymes, such as ALT and AST. Damage to the hepatobiliary system results in the release of essential enzymes, such as ALP, and/or reduces the biosynthetic and catabolic capacities of the liver. High levels of liver enzymes are signs of hepatocellular toxicity, whereas a decrease may indicate an enzyme inhibition [37, 38]. However, IP administration is less likely to be toxic to the liver than oral administration because the liver’s first-pass effect is minimized ​ [39].

In the present study, intraperitoneal administration of HTX at 50 mg/kg body weight had no effect on AST, ALT, or ALP enzyme levels. The levels of these enzymes did not significantly differ between the treated and control groups. As the primary excretory organ, the kidney is a major target of drug-induced toxicity because it is naturally exposed to a greater proportion of circulating drugs and chemicals. As BUN and creatinine are excreted primarily by the kidneys, blood chemistry analyses that measure BUN and creatinine concentrations are most commonly used to evaluate renal function. High levels of BUN and creatinine have been detected in the blood [40, 41]. The present study revealed that an acute single dose of HTX did not alter blood BUN and creatinine levels compared to the control. The liver and kidney histology were also investigated to confirm the acute toxicity of HTX. The result showed that the HTX-treated group possessed normal hepatocyte histology, including morphology, cell size, arrangement, and color staining. For kidney histopathology, HTX did not present with lesions and had normal glomeruli, Bowman’s capsules, and renal cells.

Regarding acute toxicity, biochemical markers of the liver and kidney, consistent with the results of the histopathological examination, showed that HTX did not produce any abnormalities and did not cause mortality up to a dose of 50 mg/kg body weight. These results suggest that intraperitoneal (IP) administration of HTX is relatively safe and has a median lethal dose (LD50) greater than 50 mg/kg body weight. Generally, if the LD50 of a test substance is three times greater than the minimum effective dose (MED), it is considered a good candidate for further studies [42]. However, HTX showed no lethality in mice at 50 mg/kg, which is more than three times the MED. Gross physical and behavioral observations of experimental mice revealed no visible signs of acute toxicity, indicating that HTX is safe.

Pharmacokinetic parameters aid in understanding the body’s reactions to drugs and have several uses in toxicology and biopharmaceutics [43]. The study of pharmacokinetics of compounds for malaria treatment drug development is an important step toward further evaluation in clinical trials [44]. In this study, we selected the IP route because it is easy, quick, minimally stressful for animals and suitable for our test compound, which is poorly soluble and avoids gastrointestinal tract and potential degradation of the compound [45]. The concentration-time curve of HTX showed fluctuations that may be attributed to the pharmacokinetic phenomenon of tissue distribution [43]. The calculated elimination t1/2 after the IP administration of HTX was 13.88 h. This represents a relatively long elimination half-life when compared to those of other antimalarial drugs, such as artesunate (0.8–1.2 h) [46], dihydroartemisinin (0.9–2 h [47, 48], and arterolane (2–4 h) [49]. Drugs with an elimination half-life longer than that of current antimalarial drugs may shorten the duration of malaria treatment. This would reduce the cost of current therapies [50].

Regarding malaria treatment, WHO recommends using triple artemisinin-based combination therapy (TACT), which combines short-acting artemisinin and two long-acting partner drugs [3]. The short but rapidly acting artemisinin quickly reduces the total number of parasites circulating in a patient, thus reducing the stochiastic chances for the selection of resistant mutants in the remaining low number of parasites. A longer-acting partner drug is continued suppression, complete clearance of parasites, and prevention of drug resistance [51].

Conclusions

This is the first report on the antimalarial activity of HTX in a mouse model. This study demonstrated that HTX possessed potent antimalarial activity against P. berghei. A single IP injection of HTX resulted in no behavioral changes or mortality. The LD50 of HTX was greater than 50 mg/kg body weight. Pharmacokinetics study showed that HTX has a long elimination half-life of 13.88 h. Further studies are required to elucidate the mechanism of action of HTX against malarial infections. Furthermore, this compound should be tested in combination with other antimalarial drugs to better assess its efficacy and adverse effects. This study can be considered an important step in identifying new antimalarial lead compounds to address the emergence of antimalarial drug resistance.

Data availability

All data generated or analyzed during this study are included in this published article. Additional files are available from the corresponding author on reasonable request.

Abbreviations

ACT:

Artemisinin-based combination therapy

ALP:

Alkaline phosphatase

ALT:

Alanine aminotransferase

AST:

Aspartate aminotransferase

AUC:

Area under the concentration-time curve

CC:

Column chromatography

Cmax :

Maximum concentration achieved

H E:

Hematoxylin and eosin

HTX:

1-Hydroxy-5,6,7-trimethoxyxanthone

ICR:

Institute of Cancer Research

IP:

Intraperitoneal

LC-MS/MS:

Liquid chromatography triple quadrupole mass spectrometry

LD50 :

Median lethal dose

MED:

Minimum effective dose

MRM:

Multiple reaction monitoring

MRT:

Mean residence time

NMR:

Nuclear magnetic resonance

NSS:

Normal saline solution

QCC:

Quick column chromatography

t1/2 :

Elimination half-life

Tmax :

Time to attain maximum concentration

References

  1. Alehegn AA, Yesuf JS, Birru EM. Antimalarial activity of crude extract and solvent fractions of the leaves of Bersama Abyssinica Fresen. (Melianthaceae) against Plasmodium berghei infection in Swiss albino mice. Evid Based Complement Alternat Med. 2020;2020:9467359.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Muhaimin M, Madyawati L, Riski Dwimalida P, Anis Yohana C, Andreas Yoga A, Normalita Eka P et al. Antiplasmodial activity of methanolic leaf extract of mangrove plants against Plasmodium berghei. Pharmacogn J. 2019;11(5).

  3. WHO. World malaria report 2022. Geneva: World Health Organization; 2022.

    Google Scholar 

  4. Pan WH, Xu XY, Shi N, Tsang SW, Zhang HJ. Antimalarial activity of plant metabolites. Int J Mol Sci. 2018;19(5).

  5. Koehne E, Kreidenweiss A, Adegbite BR, Manego RZ, McCall MBB, Mombo-Ngoma G, et al. In vitro activity of eravacycline, a novel synthetic halogenated tetracycline, against the malaria parasite Plasmodium Falciparum. J Glob Antimicrob Resist. 2021;24:93–7.

    Article  CAS  PubMed  Google Scholar 

  6. Fenta M, Kahaliw W. Evaluation of antimalarial activity of hydromethanolic crude extract and solvent fractions of the leaves of Nuxia congesta R. Br. Ex Fresen (Buddlejaceae) in Plasmodium Berghei infected mice. J Exp Pharmacol. 2019;11:121–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cui L, Mharakurwa S, Ndiaye D, Rathod PK, Rosenthal PJ. Antimalarial drug resistance: literature review and activities and findings of the ICEMR network. Am J Trop Med Hyg. 2015;93(3 Suppl):57–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bekono BD, Ntie-Kang F, Onguéné PA, Lifongo LL, Sippl W, Fester K, et al. The potential of anti-malarial compounds derived from African medicinal plants: a review of pharmacological evaluations from 2013 to 2019. Malar J. 2020;19(1):183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Uzor PF, Prasasty VD, Agubata CO. Natural products as sources of antimalarial drugs. Evid Based Complement Alternat Med. 2020;2020:9385125.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Kweyamba PA, Zofou D, Efange N, Assob J-CN, Kitau J, Nyindo M. In vitro and in vivo studies on anti-malarial activity of Commiphora africana and Dichrostachys cinerea used by the Maasai in Arusha region, Tanzania. Malar J. 2019;18(1):119.

  11. Waluyo D, Prabandari EE, Pramisandi A, Hidayati DN, Chrisnayanti E, Puspitasari DJ, et al. Exploring natural microbial resources for the discovery of anti-malarial compounds. Parasitol Int. 2021;85:102432.

    Article  CAS  PubMed  Google Scholar 

  12. Achan J, Talisuna AO, Erhart A, Yeka A, Tibenderana JK, Baliraine FN, et al. Quinine, an old anti-malarial drug in a modern world: role in the treatment of malaria. Malar J. 2011;10:144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Su XZ, Miller LH. The discovery of artemisinin and the Nobel Prize in physiology or medicine. Sci China Life Sci. 2015;58(11):1175–9.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Sangkaruk R, Rungrojsakul M, Tima S, Anuchapreeda S. Effect of Thai saraphi flower extracts on WT1 and Bcr/Abl protein expression in leukemic cell lines. Afr J Tradit Complement Altern Med. 2017;14(2):16–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ninomiya K, Shibatani K, Sueyoshi M, Chaipech S, Pongpiriyadacha Y, Hayakawa T, et al. Aromatase inhibitory activity of geranylated coumarins, mammeasins C and D, isolated from the flowers of Mammea siamensis. Chem Pharm Bull. 2016;64(7):880–5.

    Article  CAS  Google Scholar 

  16. Tung NH, Uto T, Sakamoto A, Hayashida Y, Hidaka Y, Morinaga O, et al. Antiproliferative and apoptotic effects of compounds from the flower of Mammea siamensis (miq.) T. Anders. On human cancer cell lines. Bioorg Med Chem Lett. 2013;23(1):158–62.

    Article  CAS  PubMed  Google Scholar 

  17. Luo F, Sugita H, Muraki K, Saeki S, Chaipech S, Pongpiriyadacha Y, et al. Anti-proliferative activities of coumarins from the Thai medicinal plant Mammea siamensis (miq.) T. Anders. Against human digestive tract carcinoma cell lines. Fitoterapia. 2021;148:104780.

    Article  CAS  PubMed  Google Scholar 

  18. Morikawa T, Sueyoshi M, Chaipech S, Matsuda H, Nomura Y, Yabe M, et al. Suppressive effects of coumarins from Mammea siamensis on inducible nitric oxide synthase expression in RAW264.7 cells. Bioorg Med Chem. 2012;20(16):4968–77.

    Article  CAS  PubMed  Google Scholar 

  19. Chaniad P, Techarang T, Phuwajaroanpong A, Horata N, Septama AW, Punsawad C. Exploring potential antimalarial candidate from medicinal plants of Kheaw Hom remedy. Trop Med Infect Dis. 2022;7(11).

  20. Chaniad P, Chukaew A, Payaka A, Phuwajaroanpong A, Techarang T, Plirat W, et al. Antimalarial potential of compounds isolated from Mammea Siamensis T. Anders. Flowers: in vitro and molecular docking studies. BMC Complement Med Ther. 2022;22(1):266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Plirat W, Chaniad P, Phuwajaroanpong A, Septama AW, Punsawad C. Phytochemical, antimalarial, and acute oral toxicity properties of selected crude extracts of Prabchompoothaweep remedy in Plasmodium berghei-infected mice. Trop Med Infect Dis. 2022;7(12):395.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Chaniad P, Techarang T, Phuwajaroanpong A, Punsawad C. Antimalarial activity and toxicological assessment of Betula alnoides extract against Plasmodium berghei infections in mice. Evid Based Complement Altern Med. 2019;2019:2324679.

    Article  Google Scholar 

  23. Peters W. The four-day suppressive in vivo antimalarial test. Ann Trop Med Parasitol. 1975;69:155–71.

    Article  CAS  PubMed  Google Scholar 

  24. OECD. Test No. 425: acute oral toxicity: up-and-down procedure2022.

  25. Wichapoon B, Punsawad C, Chaisri U, Viriyavejakul P. Glomerular changes and alterations of zonula occludens-1 in the kidneys of Plasmodium Falciparum malaria patients. Malar J. 2014;13(1):176.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Viriyavejakul P, Khachonsaksumet V, Punsawad C. Liver changes in severe Plasmodium Falciparum malaria: histopathology, apoptosis and nuclear factor kappa B expression. Malar J. 2014;13(1):106.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Patnaik MP, Sharma V, Parmar VS, Boll PM. Synthesis of new trioxygenated xanthones of Tovomita Excelsa. ChemInform. 1987;18.

  28. Remali J, Sahidin I, Aizat WM. Xanthone biosynthetic pathway in plants: a review. Front Plant Sci. 2022;13:809497.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Tovilovic Kovacevic G, Zogovic N, Krstić-Milošević D. Secondary metabolites from endangered Gentiana, Gentianella, Centaurium, and Swertia species (Gentianaceae): promising natural biotherapeutics. 2020. p. 335 – 84.

  30. Mina EC, Mina JF. Ethnobotanical survey of plants commonly used for diabetes in Tarlac of central Luzon Philippines. IIUM Med J Malays. 2017;16(1).

  31. Chatatikun M, Chiabchalard A. Thai plants with high antioxidant levels, free radical scavenging activity, anti-tyrosinase and anti-collagenase activity. BMC Complement Altern Med. 2017;17(1):487.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zhang H, Tan Y-p, Zhao L, Wang L, Fu N-j, Zheng S-p, et al. Anticancer activity of dietary xanthone α-mangostin against hepatocellular carcinoma by inhibition of STAT3 signaling via stabilization of SHP1. Cell Death Dis. 2020;11(1):63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Upegui Y, Robledo SM, Gil Romero JF, Quiñones W, Archbold R, Torres F, et al. In vivo antimalarial activity of α-mangostin and the new xanthone δ-mangostin. Phytother Res. 2015;29(8):1195–201.

    Article  CAS  PubMed  Google Scholar 

  34. Kelly JX, Winter R, Peyton DH, Hinrichs DJ, Riscoe M. Optimization of xanthones for antimalarial activity: the 3,6-bis-omega-diethylaminoalkoxyxanthone series. Antimicrob Agents Chemother. 2002;46(1):144–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Thome R, Lopes S, Costa F, Verinaud L. Chloroquine: modes of action of an undervalued drug. Immunol Lett. 2013;153.

  36. Pillat MM, Krüger A, Guimarães LMF, Lameu C, de Souza EE, Wrenger C, et al. Insights in chloroquine action: perspectives and implications in Malaria and COVID-19. Cytometry Part A. 2020;97(9):872–81.

    Article  CAS  Google Scholar 

  37. Nigatu TA, Afework M, Urga K, Ergete W, Makonnen E. Toxicological investigation of acute and chronic treatment with Gnidia Stenophylla Gilg root extract on some blood parameters and histopathology of spleen, liver and kidney in mice. BMC Res Notes. 2017;10(1):625.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ugwah-Oguejiofor CJ, Okoli CO, Ugwah MO, Umaru ML, Ogbulie CS, Mshelia HE, et al. Acute and sub-acute toxicity of aqueous extract of aerial parts of Caralluma dalzielii N. E. Brown in mice and rats. Heliyon. 2019;5(1):e01179.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Turner PV, Brabb T, Pekow C, Vasbinder MA. Administration of substances to laboratory animals: routes of administration and factors to consider. J Am Assoc Lab Anim Sci. 2011;50(5):600–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Yudhani R, Pesik R, Azzahro S, Anisa A, Hendriyani R. Renal function parameter on acute toxicity test of kapulaga (Amomum cardamom) seed extract in rat. IOP Conf Series: Mater Sci Eng. 2019;578:012053.

    Article  CAS  Google Scholar 

  41. Rivadeneyra-Domínguez E, Becerra-Contreras Y, Vázquez-Luna A, Díaz-Sobac R, Rodríguez-Landa JF. Alterations of blood chemistry, hepatic and renal function, and blood cytometry in acrylamide-treated rats. Toxicol Rep. 2018;5:1124–8.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Auletta CS. RAC M. Acute, subchronic, and chronic toxicology. Handb Toxicol. 1995:51–162.

  43. Hussain K, Ismail Z, Sadikun A, Ibrahim P. Bioactive markers based pharmacokinetic evaluation of extracts of a traditional medicinal plant, Piper Sarmentosum. Evid Based Complement Alternat Med. 2011;2011:980760.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Okada K, Sato A, Hiramoto A, Isogawa R, Kurosaki Y, Higaki K, et al. Pharmacokinetic analysis of new synthetic antimalarial N-251. Trop Med Int Health. 2019;47(1):40.

    Article  Google Scholar 

  45. Al Shoyaib A, Archie SR, Karamyan VT. Intraperitoneal route of drug administration: should it be used in experimental animal studies? Pharm Res. 2019;37(1):12.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Morris CA, Duparc S, Borghini-Fuhrer I, Jung D, Shin C-S, Fleckenstein L. Review of the clinical pharmacokinetics of artesunate and its active metabolite dihydroartemisinin following intravenous, intramuscular, oral or rectal administration. Malar J. 2011;10(1):263.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tarning J, Rijken MJ, McGready R, Phyo AP, Hanpithakpong W, Day NP, et al. Population pharmacokinetics of dihydroartemisinin and piperaquine in pregnant and nonpregnant women with uncomplicated malaria. Antimicrob Agents Chemother. 2012;56(4):1997–2007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hong X, Liu C-h, Huang X-t, Huang T-l, Ye S-m, Ou W-p, et al. Pharmacokinetics of dihydroartemisinin in Artekin tablets for single and repeated dosing in Chinese healthy volunteers. Biopharm Drug Dispos. 2008;29(4):237–44.

    Article  CAS  PubMed  Google Scholar 

  49. Gautam A, Ahmed T, Sharma P, Varshney B, Kothari M, Saha N, et al. Pharmacokinetics and pharmacodynamics of arterolane maleate following multiple oral doses in adult patients with P. Falciparum malaria. J Clin Pharmacol. 2011;51(11):1519–28.

    Article  CAS  PubMed  Google Scholar 

  50. Tanner L, Haynes RK, Wiesner L. An in vitro ADME and in vivo pharmacokinetic study of novel TB-active decoquinate derivatives. Front Pharmacol. 2019;10.

  51. Bassat Q, Maïga-Ascofaré O, May J, Clain J, Mombo-Ngoma G, Groger M, et al. Challenges in the clinical development pathway for triple and multiple drug combinations in the treatment of uncomplicated falciparum malaria. Malar J. 2022;21(1):61.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful to the staff at the Research Institute for Health Sciences, Walailak University, Thailand, for their assistance with animal experiments. Additionally, we thank the staff at the Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Thailand, for their assistance with histological preparation and staining.

Funding

This research was supported by the Thailand Science Research and Innovation Fund (contract no. FRB650082/0227-WU03). The funders had no role in the study design, data collection and analysis, decision to publish, or manuscript preparation.

Author information

Authors and Affiliations

Authors

Contributions

PC, AC, GY, and CP conceived and designed the study. PC and CP contributed to project administration and validation. PC, AC, PN, GY, LC, AP, WP, AK, and CP were responsible for methodology and investigation. AP and WP performed the statistical analyses. CP supervised the project. PC, CP, and GY drafted the manuscript. PC, AWS, and CP reviewed and edited the manuscript. All the authors have read and agreed to the final version of the manuscript.

Corresponding author

Correspondence to Chuchard Punsawad.

Ethics declarations

Ethics approval and consent to participate

This study was reviewed and approved by the Animal Ethics Committee of Walailak University, Thailand (certificate number: WU-ACUC-65015). The experimental process followed all ethical licensing agreements, guidelines and principles for animal care and use. Meanwhile, this study was performed in compliance with the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chaniad, P., Chukaew, A., Na-ek, P. et al. In vivo antimalarial effect of 1-hydroxy-5,6,7-trimethoxyxanthone isolated from Mammea siamensis T. Anders. flowers: pharmacokinetic and acute toxicity studies. BMC Complement Med Ther 24, 129 (2024). https://doi.org/10.1186/s12906-024-04427-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-024-04427-z

Keywords