Next Article in Journal
Next-Generation Heterocyclic Electrophiles as Small-Molecule Covalent MurA Inhibitors
Previous Article in Journal
Treatment of Central Nervous System Infection Caused by Multidrug-Resistant Klebsiella pneumoniae with Colistin Sulfate Intravenously and Intrathecally: A Case Report
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pharmacokinetic Study and Metabolite Identification of 1-(3′-bromophenyl)-heliamine in Rats

1
State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, CAS Key Laboratory of Chemistry of Plant Resources in Arid Regions, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
2
University of Chinese Academy of Sciences, No. 19 (A) Yuquan Road, Shijingshan District, Beijing 100049, China
3
S. Yu. Yunusov Institute of the Chemistry of Plant Substances, Academy of Sciences of the Republic of Uzbekistan, Tashkent 100170, Uzbekistan
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(12), 1483; https://doi.org/10.3390/ph15121483
Submission received: 18 October 2022 / Revised: 21 November 2022 / Accepted: 22 November 2022 / Published: 29 November 2022
(This article belongs to the Section Pharmacology)

Abstract

:
Tetrahydroisoquinolines have been widely investigated for the treatment of arrhythmias. 1−(3′−bromophenyl)−heliamine (BH), an anti−arrhythmias agent, is a synthetic tetrahydroisoquinoline. This study focuses on the pharmacokinetic characterization of BH, as well as the identification of its metabolites, both in vitro and in vivo. A UHPLC−MS/MS method was developed and validated to quantify BH in rat plasma with a linear range of 1–1000 ng/mL. The validated method was applied to a pharmacokinetic study in rats. The maximum concentration Cmax (568.65 ± 122.14 ng/mL) reached 1.00 ± 0.45 h after oral administration. The main metabolic pathways appeared to be phase-I of demethylation, dehydrogenation, and epoxidation, and phase II of glucuronide and sulfate metabolites. Finally, a total of 18 metabolites were characterized, including 10 phase I metabolites and 8 phase II metabolites. Through the above studies, we have gained a better understanding of the absorption and metabolism of BH in vitro and in vivo, which will provide us with guidance for future in-depth studies on this compound.

Graphical Abstract

1. Introduction

Cardiovascular disease, a widespread condition with 80% of the disease burden all over the world, includes arrhythmia, ischemic heart disease, chronic rheumatic heart disease, acute rheumatic fever, pulmonary embolism, heart failure, and hypertension [1,2,3,4]. It is estimated that 17.9 million people die each year from cardiovascular diseases worldwide, and this number is continuously increasing [5,6]. Worldwide, low and middle-income countries, especially China, have the highest burden of cardiovascular diseases [7]. According to the statistics [2], 330 million people suffer from cardiovascular diseases in China, including 4.87 million with arrhythmia. In 2019 alone, the cost of hospitalization for cardiovascular disease in China was CNY 177.338 billion, of which CNY 18.099 billion was spent on arrhythmia [2,8,9]. Currently, antiarrhythmic medicines are clinically classified into four classes, according to their mechanisms [10]: Category I: sodium channel blockers (quinidine, lidocaine, and propafenone) [11]; Category II: β-blockers (propranolol); Category III: anti-fibrillation drugs blocking K+ channels (amiodarone); and Category IV: Ca2+ channel blockers (verapamil and diltiazem) [12].
Tetrahydroisoquinolines are structurally novel calcium channel blockers, similar to dihydropyridines, that act mainly at the dihydropyridine site of the cellular L-type calcium channel [13,14]. Several tetrahydroisoquinoline derivatives, including those from natural and synthetic products, have shown excellent antiarrhythmic effects. For example, tetrandrine [15], berberine [16,17], dauricine [17,18], protopine [19], and other natural products have excellent anti−arrhythmic activity. 1−(3′−bromophenyl)−heliamine (BH) (Figure 1) is a synthetic tetrahydroisoquinoline alkaloid [20]. However, the possible anti-arrhythmic mechanism of BH is as a membrane surface Na+–Ca2+ exchange inhibitor [21]. In addition, it reduces sarcoplasmic reticulum Ca2+ release by inhibiting sarcoplasmic reticulum RyR2Ca2+ channels [22]. BH is still undergoing preclinical studies, and its pharmacokinetic profile and metabolites have not yet been studied. Therefore, it is necessary to investigate the pharmacokinetics and metabolites of BH in rats.
The aim of this study is to investigate the absolute bioavailability of BH in rats by both oral and intravenous administration. Subsequently, the metabolites of BH were investigated by in silico, in vitro, and in vivo studies: (a) the in silico prediction of possible metabolic sites of BH by simulating calculation; (b) the in vitro metabolites of BH in rat liver microsomes; (c) the in vivo metabolites of BH in plasma, urine, and feces. The metabolic pathways of BH in rats were proposed, and the pharmacokinetic parameters of BH in rats were summarized.

2. Results and Discussion

2.1. Pharmacokinetic (PK) Analysis and PK Parameter

The mean plasma concentration profiles of BH after oral and injection administration are shown in Figure 2. The pharmacokinetic parameters obtained by the non-compartmental analysis are summarized in Table 1. The Cmax of 568.65 ± 122.14 ng/mL was achieved at 1.00 ± 0.45 h after oral administration, indicating that BH was quickly absorbed into the blood circulatory system. The T1/2 was 1.62 ± 0.18 h after oral administration, indicating that BH was cleared rapidly from the blood circulatory system. The AUC0-t and AUC0-inf values after oral administration were 1931.81 ± 492.08 and 1968.64 ± 479.81 h*(ng/mL), indicating that the monitoring time was appropriate. The apparent volume of distribution was 2484.43 ± 622.32 mL, indicating a lower plasma protein binding for BH [23]. The absolute bioavailability of BH was 10.6%, indicating low system exposure.

2.2. Metabolites of BH

It is crucial for early-stage drug development to conduct metabolic profiling on a new chemical entity. Preliminary in silico metabolism calculations were performed in order to predict possible metabolic sites by XenoSite [24]. Then, in vitro and in vivo metabolites of BH were identified.

2.2.1. Metabolic Sites Prediction of BH

Traditional drug discovery takes 4–5 years, and costs more than USD 200 million for one new drug [25,26]. Even though in silico applications remain limited by shortcomings such as inconsistent and erroneous data, they help us to approach metabolic sites with a higher probability [27]. Figure 3 shows the most likely metabolic sites of BH in human liver microsomes. Figure 3A shows that the tetrahydroisoquinoline part was prone to be hydroxylated. The methoxy part was also highly susceptible to being demethylated. Figure 3B indicates the potential site of epoxidation at the benzene ring. Figure 3C shows the probability of phase II glucuronidation occurring. Figure 3D shows the probability of phase II glucuronidation after demethylation.

2.2.2. Mass Fragmentation of BH

BH is the parent compound of all its metabolites. Since the metabolites derive from BH, the mass spectral characteristics of metabolites show high correlations with those of BH. Therefore, fragmentation of BH has the highest priority in the identification study of metabolites by mass spectrometry. BH possesses a bromine atom, and the natural abundance ratio of 79Br and 81Br is 1:1. Therefore, either of the two can be selected for further analysis. Herein, 81Br was selected for elaboration. The protonated molecular ion peak of BH in ESI positive mode was m/z 350.0571 (C17H1981BrNO2). The major fragments were m/z 333.0304, 252.1145, 179.0943, 170.9628, and 164.0706. The most abundant fragment ion at m/z 179.0943 was produced by the loss of a methyl radical and the 3’-bromoband benzene. By further loss of ·CH3 (15), the fragment ion m/z 164.0706 was yielded. In addition, the C-N bond in the tetrahydroisoquinoline ring was readily cleaved. The fragment ion m/z 333.0304 was produced by the loss of NH3 (17) from the parent ion. This fragmental ion further generated a daughter ion at m/z 252.1145 by loss of a bromine radical. The fragment ion m/z 170.9628 was a ring cleavage product. Figure 4 shows the cleavage rule of BH.

2.2.3. Metabolites of BH

Accurate mass measurements and key fragmental ions of the metabolites of BH in vitro are shown in Table 2. In total, 18 metabolites were characterized, including 10 phase I and 8 phase II metabolites. The surmised pathways of metabolism suggested that BH mainly underwent desaturation, oxidation, dehydrogenation, sulfation, and glucuronidation. The fragmentation processes of M1−A/M1−B, M5, and M6 are shown in Figure 5. Since the isomers of M1−A and M1−B showed the same MS/MS fragment ions one of them was used for illustration in the following structure analysis process. This also applied to M8−A/M8−B, M9−A/M9−B, and M10−A/M10−B. Their accurate mass measurements and key fragmental ions are shown in Table 2. The accurate mass measurements and key fragmental ions of other metabolites not mentioned in the context are also shown in Table 2.
M1−A/M1−B was found to be a demethylated metabolite of BH. It showed a protonated molecular ion peak at m/z 336.0416, and its major fragmental ions were observed at m/z 319.0154, 286.9890, 238.0988, 170.9628, and 165.0788. The fragment ion m/z 170.9628 was a ring cleavage product. The most abundant fragmental ion at m/z 165.0788 was generated from the parent ion by the loss of a methyl radical and the 3’-bromoband benzene. In addition, the loss of a NH3 from the parent ion led to the fragmental ion m/z 319.0154. This fragmental ion further produced daughter ions at m/z 238.0988 and 286.9890 by loss of a bromine radical and a methanol moiety, respectively.
M5 was observed with a protonated molecular ion at m/z 322.0259. Its major product ion occurred at m/z 304.9995, 224.0830, 170.9627, and 151.0628. The fragment of m/z 304.9995 was produced from the parent ion by the loss of a NH3. It further generated two fragmental ions at m/z 224.0830 and 170.9627. The fragment at m/z 151.0628 was a ring cleavage product.
M8−A/M8−B was the sulfated metabolite of demethylated BH. It displayed the protonated molecular ion m/z 415.9984. The major fragment ions were observed at m/z at 336.0416, 319.0148, 286.9890, 238.0987, 170.9627, and 165.0787. The fragmental ion at m/z 336.0415 was produced by the loss of sulfate moiety from the parent ion. Afterward, the fragmentations were the same as those of M1−A/M1−B. Similarly, M10−A/M10−B was a glucuronidated metabolite of M5.
M6 gave a precursor ion at m/z 364.0370. Its major product ions occurred at m/z 190.0863, 175.0628, and 146.0601. By analyzing the base peak m/z 190.0863, the position of epoxidation in the bromobenzene ring was inferred. This assumption was the same as that in in silico prediction (see Figure 3B). The product ion m/z 175.0628 was 15 Da less than product ion m/z 190.0863, suggesting the subtraction of one methyl from m/z 190.0863. The product ion m/z 146.0601 was generated from ion m/z 175.0628 by the loss of a methoxy as well as electronic rearrangement.
M9−A/M9−B was a multiple-site metabolite of BH. The protonated molecular ion peak m/z 508.0437 was observed. Its major fragment ions were m/z 332.0103 and 316.9867, and the fragment ion m/z 332.0103 was formed by the loss of a glucuronosyl. This fragmental ion further produced daughter ion at m/z 316.9867 by loss of a methyl radical.
The possible metabolic pathways of BH are proposed in Figure 6. These pathways included desaturation, dehydrogenation, oxidation, epoxidation, sulfation, and glucuronidation. Notably, glucuronidated metabolites were found in urine and feces, but not in plasma.

3. Materials and Methods

3.1. Reagents and Chemicals

BH (97.5%) was obtained from S. Yu. Yunusov Institute of the Chemistry of Plant Substances, Academy of Sciences of the Republic of Uzbekistan (Tashkent, Uzbekistan). Colchicine (93.0%) (Figure 1) was supplied by the National Institute for Food and Shanghai Hong Yong Biotechnology company (Shanghai, China). The UHPLC-MS/MS grades of methanol, acetonitrile, and formic acid were procured from Thermo Fisher Scientific (Bremen, Germany). Blank rat plasma was supplied by the Wuhan purity biotechnology company (Wuhan, China). Ethanol was purchased from the Tianjin Xin platinum chemical company (Tianjin, China). Distilled water was obtained from Watsons (Guangzhou, China). Kolliphor was provided by Sigma-Aldrich (St. Louis, MO, USA). Phase I and UGT metabolic stability kits were purchased from the Beijing Hui Zhi He Yuan biotechnology company (Beijing, China).

3.2. Animals and Experiments

All twelve male Sprague–Dawley (SD) rats (weight 200 ± 20 g) were obtained from the Comparative Medicine Center of Xinjiang Medical University (Xinjiang, China).
BH was formulated in a solution of 2% ethanol, 8% Kolliphor, and 90% distilled water. The twelve rats were fasted for 12 h, but had free access to water, and were randomly divided into two groups. In the oral group, 19.2 mg/kg was orally administered to each rat. Blood was collected from the ophthalmic veins (approximately 0.3 mL) at 0, 0.25, 0.5, 1, 1.5, 2, 4, 8, 12, and 24 h for oral study. In the intravenous group, 1.9 mg/kg was administered by intravenous injection to each rat via the tail. Blood was collected from the ophthalmic veins (approximately 0.3 mL) at 0, 0.1, 0.25, 0.5, 0.75, 1, 1.5, 3.4, 7.5, 12, and 24 h. Then, samples were placed into 1.5 mL polyethylene tubes containing EDTA. Additionally, the blood samples were centrifuged (14,000 rpm) for 10 min. After centrifugation, the supernatant was extracted and then placed at −80 °C for the next step of the analysis.

3.3. Calibration Standard and Quality Control Samples in Rat Plasma

Stock standard solutions of BH (1.0 mg/mL) were prepared by dissolving in 0.1 mL of dimethyl sulfoxide and diluting with 30% acetonitrile in water. The first group of stock solution was diluted stepwise into 10, 30, 50, 100, 300, 500, 1000, 3000, 5000, and 10,000 ng/mL of BH calibrator solution. Another set of BH working solutions, with concentrations of 10, 100, 1000, and 5000 ng/mL, were used in the same way for the preparation of quality control samples. The working solution of the internal standard (1.0 mg/mL) was prepared by diluting it with 30% acetonitrile in water. All solutions were stored at 4 °C.
To prepare the calibration standards, the samples were vortexed for 3 s (3000 rpm) with an aliquot of 10 µL BH working solutions in 100 µL blank plasma. Then, 10 µL internal standard was added, and the mixture was vortexed for 3 s (3000 rpm). The mixture was vortexed for 1 min (3000 rpm); afterward, 300 µL acetonitrile was added in order to precipitate the protein. Then, the mixture was centrifuged at 14,000 rpm for 7 min. The supernatant was collected and dried with 40 °C nitrogen. The samples were redissolved with 100 µL of 28% acetonitrile in water (v/v) and sonicated for 1 min, centrifuged for 7 min, then subjected to UHPLC-MS/MS analysis. Thus, the range of the calibration curve was 1–1000 ng/mL, and the internal standard concentration was 300 ng/mL. In the same manner as described above, quality controls were prepared at four concentration levels. The concentrations of BH and internal standard were 1, 10, 100, 500, and 300 ng/mL.

3.4. Plasma Sample Preparation

A 10 µL volume of 30% acetonitrile in water (v/v) was added to 100 µL of plasma samples and vortexed for 3 s (3000 rpm). Then, 10 µL internal standard was added to mixtures and vortexed for 3 s (3000 rpm). Subsequently, samples were vortexed for 1 min, after which 300 µL acetonitrile was added for protein precipitation. Additionally, the mixtures were centrifuged for 7 min. The supernatant was collected and dried with 40 °C nitrogen. The samples were re-dissolved with 100 µL of 30% acetonitrile in water (v/v) and sonicated for 1 min, centrifuged for 7 min, and then subjected to UHPLCMS/MS analysis.

3.5. Instruments and UHPLC-MS/MS Conditions

The UHPLC-MS/MS bioanalytical method was established on a Dionex UltiMate 3000 RSL CnanoSystem coupled with Thermo Scientific Q Exactive Plus Orbitrap. The system was controlled by Xcalibur, with data processed using Thermo Xcalibur 4.2.28.14 Quan Browser (Thermo Fisher Scientific, USA).
Chromatographic separation was performed on an Acquity UHPLC BEH C18 (2.1 × 75 mm, 1.7 µm). The column temperature was set at 35 °C. The mobile phase consisted of isocratic formic acid (0.1%): acetonitrile (70:28, v/v) at a flow rate of 0.2 mL/min. The injection volume was 10 µL, and the autosampler temperature was set at 10 °C. The BH and internal standard were detected by the parallel reaction monitor (PRM) mode. The electrospray ionization (ESI) source parameters were adjusted as follows: automatic gain control (AGC) target, 1 × 106; maximum IT, 100 ms; spray voltage, 3.2 kV; capillary temp, 320 °C S–lens RF level, 55; and high collision dissociation cell (HCD) energy, 35 (for BH) and 38 (for the internal standard).
The UHPLC−MS/MS system and column were the same as those used above. The mobile phase consisted of formic acid (0.1%) and acetonitrile at a flow rate of 0.2 mL/min. The column temperature was set at 35 °C and the injection volume was 10 µL. The gradient elutions were: 0–3 min (5%B), 3–33 min (25%B), 33–60 min (60%B). The metabolites were detected by the full MS/dd MS2 mode, with a resolution of 17,500. The heated electrospray ionization (HESI) source parameters were as follows: AGC target, 1 × 106; maximum IT, 100 ms; spray voltage, 3.2 kV; capillary temp, 320 °C; S–lens RF level, 55; and HCD Energy, 20, 40, and 60.

3.6. Method Validation

3.6.1. Specificity

The selectivity was evaluated by analyzing blank plasma from six individuals, blank plasma spiked with BH and internal standard, and pharmacokinetic study samples. Blank plasma samples did not exhibit interfering peaks at the retention time of BH or the internal standard. The results are included in Figure S1.

3.6.2. Calibration Curves

A plot of the peak area ratio of BH (y) to the internal standard of the nominal concentration (x) was used to evaluate linearity. The linear range was 1–1000 ng/mL. Additionally, all calibration curves were linear using the least squares regression of the 1/x2 weighting factor. The coefficient of determination (R2) value was 0.998 for all runs, indicating an adequate linear fit. Data were processed using Thermo Xcalibur 4.2.28.14 Quan Browser.

3.6.3. Accuracy and Precision

Three consecutive batches were determined both on the same day and on three different days to validate intra- and inter-day precision and accuracy. To evaluate the accuracy and precision, four QC concentrations (1, 10 100, and 500 ng/mL) were processed on the same day (intra-day) and on three consecutive days (inter-day). The calculation of relative errors (RE) and relative standard deviations (RSD) was used to determine accuracy and precision. Finally, the intra-day accuracies were in the range of 92.34–105.80%, and the inter-day accuracies were within the range of 96.73–108.20%. The intra-day and inter-day precision values were less than 10%. The results are included in Table S1.

3.6.4. Recovery and Matrix Effect

The recovery of BH was determined at QC levels in replicates of six. A ratio of BH peak areas from extracted QC samples and blank plasma spiked with authentic standards was calculated. By comparing BH peak areas from spike-after-extraction samples to those from standard solutions at QC levels, the matrix effect was investigated. The recovery and matrix effect of the internal standard was evaluated by the same method at a concentration of 300 ng/mL. The results are included in Table S2. The recovery and matrix effects of BH from plasma were 93.35–97.13% and 100.01–107.08%, respectively.

3.6.5. Stability

Three QC concentrations (10, 100, and 500 ng/mL) in plasma samples were tested for stability under four conditions: (1) at autosampler for 4, 8, and 12 h; (2) at −80 °C for 1, 7, and 15 days; (3) at room temperature for 12 h; and (4) over three freeze–thaw cycles. In addition, the stability values of BH and internal standard in stock solution were evaluated at 4 °C for 15 days. BH was stable under the above condition. The results are included in Table S3.

3.7. Metabolic Study

3.7.1. In Silico Metabolism Calculation

The output is displayed as a color scale gradient, distributed on a scale from 0 to 1 (a value of 1 indicates the maximum level of confidence a model can have in predicting the probability that the molecule is a biotransformation site). The metabolic export of CYP is shown in the synopsis form of various human cytochrome isoenzymes (1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4). The web-based tool XenoSite was used to assess the metabolism of BH by predicting the atomic sites where BH may be modified by (i) cytochrome P450 [23], (ii) epoxidation [28], and (iii) metabolic modification catalyzed by uridine diphosphate glucuronosyltransferases (UGTs) [29]. XenoSite Web integrates machine learning from neural networks with the computation of several quantitative descriptors of molecules, including their topological and quantum chemical descriptions, as well as the reactivity of atomic positions predicted by Smart-Cyp software [30,31]. The epoxidation precursor unit was developed from the above model, which was applied to the intrinsic reactivity of different molecules with glutathione.
The XenoSite result can be interpreted as a probability that reflects both the model’s confidence about a specific fraction being metabolized and the statistical likelihood that this prediction is correct. In other words, an atom with a prediction score of 0.8 has an 80% chance of becoming an experimentally verified SOM. The output is displayed as a color scale, distributed on a scale from 0 to 1 (the value of 1 reflects the maximum confidence of the model in predicting the probability that the molecule is a biotransformation site). Human cytochrome isozymes (1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4) were added to produce the output of CYP metabolism.

3.7.2. In Vitro and In Vivo Experiments

A rat liver microsome was used the in vitro experiment. For phase I metabolism, the liver microsomes kit contained most of the phase I enzymes. In studies with liver microsomes, the in vitro metabolic system can be reconstituted if the corresponding cofactor, NADPH, is added. Then, phase I metabolic stability studies were performed by the in vitro warm incubation method. The Phase I metabolic reaction operation is as follows: (1) melt all components of the kit in an ice bath and place on ice for use. (2) Mix all components—Liquid A (NADP+, glucose 6-phosphate, and magnesium chloride, (50 μL)), Liquid B (glucose 6-phosphate dehydrogenase and sodium citrate (10 μL)), BH (10 μL), and 0.1M phosphate buffer saline (905 μL))of the incubation system and pre-incubate at 37 °C for 5 min. (3) Add the above mixture (975 μL) to a 2 mL centrifuge tube, preserve the heat in a 37 °C water bath, and then add 25 μL of liver microsome. Blow and suck three times, and mix well to start the metabolic reaction under the conditions of the 37 °C water bath. (4) After 40 min, add 1 mL of precooled acetonitrile to the incubation system to terminate the reaction.
The Phase II metabolic reaction operation is as follows: (1) melt all components of the kit in an ice bath and place on ice for use. (2) Mix all components—Liquid A (NADP+, glucose 6-phosphate, and magnesium chloride, (50 μL)), Liquid B (glucose 6-phosphate dehydrogenase and sodium citrate (10 μL)), UDPGA (50 mM, 100 μL), Alamethicin (250 μg/mL, 100 μL), d-Saccharic acid 1,4-lactone monohydrate (50 mM, 100 μL), BH (10 μL), and 0.1 M phosphate buffer saline (605 μL))—of the incubation system. (3) Add the above mixture (975 μL) into a 2 mL centrifuge tube, preserve the heat in a 37 °C water bath, and then add 25 μL of liver microsome. Blow and suck three times, and mix well to start the metabolic reaction under the conditions of the 37 °C water bath. (4) After 40 min, add 1 mL of precooled acetonitrile to the incubation system to terminate the reaction.
For the plasma study, plasma samples were prepared in the same manner as in the pharmacokinetic study, except that the re-dissolved solvent was methanol. For the urine study, the urine was collected at 0–24 and 24–48 h after administration. Urine from each period was mixed into 10 mL units, and 30 mL of acetonitrile was added. The urine samples were then sonicated for 10 min and aliquoted to 1.5 mL centrifuge tubes. The samples were vortexed for 1 min and centrifuged for 10 min (14,000 rpm), and then the supernatants were merged and dried with nitrogen at 40 °C. The samples were re-dissolved with 100 µL methanol, sonicated for 1 min, centrifuged (14,000 rpm) for 7 min, and then subjected to UHPLC-MS/MS analysis.
For the feces study, the feces were collected at 0–24 and 24–48 h after administration. The samples were dried and de-combined with 8 mL of sample and 40 mL of acetonitrile in a 50 mL centrifuge tube. Then, the samples were sonicated for 30 min. The supernatant was centrifuged at 14,000 rpm for 10 min and blow dried at 40 °C. The samples were re-dissolved with 100 µL methanol, sonicated for 1 min, centrifuged (14,000 rpm) for 7 min, then subjected to UHPLC-MS/MS analysis.

4. Conclusions

In conclusion, a composite strategy was proposed to comparatively study the pharmacokinetics of BH. Through the preclinical study, the PK properties of BH were characterized for the first time, which provided helpful information for future clinical research. The mass fragmentation rules of BH were summarized and successfully used to guide the identification of 18 metabolites, both in vitro and in vivo. The metabolic pathways of BH were proposed, including demethylation, dehydrogenation, epoxidation, sulfation, and glucuronidation. Because the in silico prediction can only supply the possible metabolic sites by human metabolic enzymes, it has some limits, even though the metabolic enzymes of rats and humans show some homology. Notably, a high probability of glucuronidation at the “N” site by human metabolic enzymes was predicted by in silico calculations. However, glucuronide metabolite at the “N” site was not found in either the rat (in vivo) study or the rat liver microsome (in vitro) study. This indicates that special attention must be paid to the metabolic differences between humans and rats in future clinical studies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15121483/s1, Figure S1: PRM extracted the ion chromatogram of 1–(3′–bromophenyl)–heliamine and colchicine in rat plasma: blank rat plasma (A); blank rat plasma spiked with 100 ng/mL and internal standard (B); an incurred sample after oral administration of 1–(3′–bromophenyl)–heliamine at 1.5 h (C); and an incurred sample after intravenous administration of 1–(3′–bromophenyl)–heliamine at 0.5 h (D). Table S1: Validation parameters: intra–day and inter–day accuracies and precisions of 1–(3′–bromophenyl)–heliamine in rat plasma (n = 6); Table S2: The validation parameters: recovery and matrix effect of 1–(3′–bromophenyl)–heliamine and the internal standard in rat plasma; Table S3: Stability results of the validation parameters under different conditions.

Author Contributions

Conceptualization, Y.L. and R.X.; methodology, R.X., Y.L., M.Z., and R.A.; software, Y.L., R.X., and Z.S.; validation, Y.L., M.H., and R.A.; formal analysis, R.X., Y.L., R.A., M.H., and V.V.I.; investigation, R.X. and M.Z.; resources, R.A., Z.S., V.V.I., and M.Z.; data curation, R.X. and Y.L.; writing—original draft preparation, R.X.; writing—review and editing, Y.L.; visualization, R.X.; supervision, Y.L. and M.H.; project administration, Y.L. and M.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Key R&D Program of China (Grant No. 2021YFE0104000), the Xinjiang Uygur Autonomous Region Regional Collaborative Innovation Special Project (International Cooperation Project, Project No. 2022E01007), and the National Key R&D Program of China under Grant 2020YFE0205600.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Review Board of Faculty of Medicine, Xinjiang Medical University, China (protocol code was IACUC-20200924-29 and date of approval was 24 September 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained in the article and Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dehghan, M.; Mente, A.; Zhang, X.; Swaminathan, S.; Li, W.; Mohan, V.; Iqbal, R.; Kumar, R.; Wentzel-Viljoen, E.; Rosengren, A.; et al. Associations of fats and carbohydrate intake with cardiovascular disease and mortality in 18 countries from five continents (PURE): A prospective cohort study. Lancet 2017, 390, 2050–2062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Report on cardiovascular health and diseases in China 2021: An updated summary. Chin. Health Econ. 2022, 37, 533–578.
  3. Shi, Y.; Zhang, H.; Huang, S.; Yin, L.; Wang, F.; Luo, P.; Huang, H. Epigenetic regulation in cardiovascular disease: Mechanisms and advances in clinical trials. Signal Transduct. Target. Ther. 2022, 7, 1–28. [Google Scholar] [CrossRef]
  4. Jacob, V.; Reynolds, J.A.; Chattopadhyay, S.K.; Hopkins, D.P.; Therrien, N.L.; Jones, C.D.; Durthaler, J.M.; Rask, K.J.; Cuellar, A.E.; Clymer, J.M.; et al. Pharmacist interventions for medication adherence: Community guide economic reviews for cardiovascular disease. Am. J. Prev. Med. 2022, 62, E202–E222. [Google Scholar] [CrossRef]
  5. Roth, G.A.; Mensah, G.A.; Fuster, V. The global burden of cardiovascular diseases and risks. J. Am. Coll. Cardiol. 2020, 76, 2980–2981. [Google Scholar] [CrossRef]
  6. World Health Organization. World Health Statistics 2022: Monitoring Health for the SDGs, Sustainable Development Goals. Available online: https://www.who.int/publications/i/item/9789240027053 (accessed on 10 October 2022).
  7. Yi, T.W.; Levin, A. Sex, gender, and cardiovascular disease in chronic kidney disease. Semin. Nephrol. 2022, 42, 197–207. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, Y.; Zhai, T.; Chai, P.; Hwang, Y.; Wan, Q. Study on accounting and projection of curative expenditure on cardiovascular and cerebrovascular diseases in China. Chin. Health Econ. 2019, 38, 18–22. [Google Scholar]
  9. Shaw, L.J.; Goyal, A.; Mehta, C.; Xie, J.; Phillips, L.; Kelkar, A.; Knapper, J.; Berman, D.S.; Nasir, K.; Veledar, E.; et al. 10-Year resource utilization and costs for cardiovascular care. J. Am. Coll. Cardiol. 2018, 71, 1078–1089. [Google Scholar] [CrossRef]
  10. Hashimoto, K. Arrhythmia models for drug research: Classification of antiarrhythmic drugs. J. Pharmacol. Sci. 2007, 103, 333–346. [Google Scholar] [CrossRef] [Green Version]
  11. Langenfeld, H.; Weirich, J.; Köhler, C.; Kochsiek, K. Atrioventricular differences in the class I effect of antiarrhythmic drugs (prajmaline,propafenone,quindine). Eur. J. Pharmacol. 1990, 183, 1163. [Google Scholar] [CrossRef]
  12. Al-Hashimi, M.; Thompson, J.P. Drugs acting on the heart: Anti-arrhythmics. Anaesth. Intensive Care Med. 2012, 13, 374–377. [Google Scholar] [CrossRef] [Green Version]
  13. Dong, H.; Lee, C.M.; Huang, W.L.; Peng, S.X. World Health Organization cardiovascular effects of substituted tetrahydroisoquinolines in rats. Br. J. Pharmacol. 1992, 107, 262–268. [Google Scholar] [CrossRef] [PubMed]
  14. Dong, H.; Waldron, G.J.; Cole, W.C.; Triggle, C.R. Cardiovascular pharmacology of CPU-23: A novel calcium channel blocker. Cardiovasc. Drug Rev. 1996, 14, 364–379. [Google Scholar] [CrossRef]
  15. Wu, S.; Yu, X.; Shan, J.; Wong, T.; Chen, C.; Pang, K. Cardiac effects of the extract and active components of radix stephaniae tetrandrae I. Electrically-induced intracellular calcium transient and protein release during the calcium paradox. Life Sci. 2001, 68, 2853–2861. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, L.-H.; Li, X.-L.; Li, Q.; Fu, Y.; Yu, H.-J.; Sun, Y.-Q.; Zhang, L.; Shan, H.-L. Berberine alleviates ischemic arrhythmias via recovering depressed Ito and ICa currents in diabetic rats. Phytomedicine 2012, 19, 206–210. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, Y.X.; Zheng, Y.M. Ionic mechanism responsible for prolongation of cardiac action-potential duration by berberine. J. Cardiovasc. Pharmacol. 1997, 30, 214–222. [Google Scholar] [CrossRef]
  18. Zhu, J.Q.; Zeng, F.D.; Hu, C.J. Prothcive and antiarrhythmic effects of dauricine and verapamil on acute myocardial-infarction in anesthetized dogs. Acta Pharmacol. Sin. 1992, 13, 249–251. [Google Scholar]
  19. Song, L.-S.; Ren, G.-J.; Chen, Z.-L.; Chen, Z.-H.; Zhou, Z.-N.; Cheng, H. Electrophysiological effects of protopine in cardiac myocytes: Inhibition of multiple cation channel currents. Br. J. Pharmacol. 2000, 129, 893–900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Zhurakulov, S.N.; Vinogradova, V.I.; Levkovich, M.G. Synthesis of 1-aryltetrahydroisoquinoline alkaloids and their analogs. Chem. Nat. Compd. 2013, 49, 70–74. [Google Scholar] [CrossRef]
  21. Jumaev, I.Z.; Jurakulov, S.N.; Usmanov, P.B. The inotropic effect of 1-(4-dimetylaminophenyl)-6,7-dimetoxy-1,2,3,4-tetrahydroisoquinoline (F24). Pharm. J. 2018, 4, 93–97. [Google Scholar]
  22. Jumayev, I.; Usmanov, P.; Rustamov, S.; Zhurakulov, S. Comparative inotropic effects of the some isoquinoline alkaloids. Biomed. Pharmacother. 2020, 13, 325–333. [Google Scholar] [CrossRef]
  23. Davies, B.; Morris, T. Physiological-parameters in laboratory-animals and humans. Pharm. Res. 1993, 10, 1093–1095. [Google Scholar] [CrossRef] [PubMed]
  24. Zaretzki, J.; Matlock, M.; Swamidass, S.J. Xenosite: Accurately predicting CYP-mediated sites of metabolism with neural networks. J. Chem. Inf. Model. 2013, 53, 3373–3383. [Google Scholar] [CrossRef] [PubMed]
  25. DiMasi, J.A.; Grabowski, H.G.; Hansen, R.W. Innovation in the pharmaceutical industry: New estimates of R&D costs. J. Health Econ. 2016, 47, 20–33. [Google Scholar]
  26. Paul, S.M.; Mytelka, D.S.; Dunwiddie, C.T.; Persinger, C.C.; Munos, B.H.; Lindborg, S.R.; Schacht, A.L. How to improve R&D productivity: The pharmaceutical industry’s grand challenge. Nat. Rev. Drug Discov. 2010, 9, 203–214. [Google Scholar] [PubMed]
  27. Kazmi, S.R.; Jun, R.; Yu, M.-S.; Jung, C.; Na, D. In silico approaches and tools for the prediction of drug metabolism and fate: A review. Comput. Biol. Med. 2019, 106, 54–64. [Google Scholar] [CrossRef] [PubMed]
  28. Hughes, T.B.; Miller, G.P.; Swamidass, S.J. Modeling epoxidation of drug-like molecules with a deep machine learning network. ACS Cent. Sci. 2015, 1, 168–180. [Google Scholar] [CrossRef] [PubMed]
  29. Dang, N.L.; Hughes, T.B.; Krishnamurthy, V.; Swamidass, S.J. A simple model predicts UGT-mediated metabolism. Bioinformatics 2016, 32, 3183–3189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Rydberg, P.; Gloriam, D.E.; Zaretzki, J.; Breneman, C.; Olsen, L. Oxidative medicine and cellular longevity SMARTCyp: A2D method for prediction of cytochrome P450-mediated drug metabolism. ACS Med. Chem. Lett. 2010, 1, 96–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Zaretzki, J.; Rydberg, P.; Bergeron, C.; Bennett, K.P.; Olsen, L.; Breneman, C.M. RS-predictor models augmented with SMARTCyp reactivities: Robust metabolic regioselectivity predictions for nine CYP Isozymes. J. Chem. Inf. Model. 2012, 52, 1637–1659. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of 1−(3′−bromophenyl)−heliamine and colchicine (internal standard).
Figure 1. Chemical structures of 1−(3′−bromophenyl)−heliamine and colchicine (internal standard).
Pharmaceuticals 15 01483 g001
Figure 2. Mean blood concentration vs. time profiles of 1−(3′−bromophenyl)−heliamine after administration to SD rats (n = 6), (A) 19.2 mg/kg, orally; (B) 1.9 mg/kg, intravenously.
Figure 2. Mean blood concentration vs. time profiles of 1−(3′−bromophenyl)−heliamine after administration to SD rats (n = 6), (A) 19.2 mg/kg, orally; (B) 1.9 mg/kg, intravenously.
Pharmaceuticals 15 01483 g002
Figure 3. In silico prediction of (A) sites of metabolism in human liver microsomes; (B) site of epoxidation; (C) site of glucuronidation in 1−(3′−bromophenyl)−heliamine; (D) site of glucuronidation in O-demethylated metabolite. Potential sites of metabolism are highlighted by a colour gradient, distributed throughout a range of 0 (no colour; minimal probability) to 1 (red colour; maximal probability).
Figure 3. In silico prediction of (A) sites of metabolism in human liver microsomes; (B) site of epoxidation; (C) site of glucuronidation in 1−(3′−bromophenyl)−heliamine; (D) site of glucuronidation in O-demethylated metabolite. Potential sites of metabolism are highlighted by a colour gradient, distributed throughout a range of 0 (no colour; minimal probability) to 1 (red colour; maximal probability).
Pharmaceuticals 15 01483 g003
Figure 4. The MS/MS spectra of 1−(3′−bromophenyl)−heliamine.
Figure 4. The MS/MS spectra of 1−(3′−bromophenyl)−heliamine.
Pharmaceuticals 15 01483 g004
Figure 5. The MS/MS spectra of M1−A/M1−B, M5, and M6.
Figure 5. The MS/MS spectra of M1−A/M1−B, M5, and M6.
Pharmaceuticals 15 01483 g005
Figure 6. The proposed metabolic pathways of 1−(3′−bromophenyl)−heliamine.
Figure 6. The proposed metabolic pathways of 1−(3′−bromophenyl)−heliamine.
Pharmaceuticals 15 01483 g006
Table 1. The main pharmacokinetic parameters of 1−(3′−bromophenyl)−heliamine in rats after intravenous and oral administration (n = 6, mean ± SD).
Table 1. The main pharmacokinetic parameters of 1−(3′−bromophenyl)−heliamine in rats after intravenous and oral administration (n = 6, mean ± SD).
ParametersUnitPO (19.2 mg/kg)IV (1.9 mg/kg)
AUC0–t *h·(ng/mL)1931.81 ± 492.081810.30 ± 696.02
AUC0–inf *h·(ng/mL)1968.64 ± 479.811902.34 ± 664.42
MRT0–t *h2.67 ± 0.381.95 ± 0.62
MRT0–inf *h2.85 ± 0.242.47 ± 0.86
Tmax *h1.00 ± 0.45
T1/2 *h1.62 ± 0.182.08 ± 1.01
Cmax *ng/mL568.65 ± 122.14905.63 ± 46.99
CL *mL/h1056.47 ± 202.07117.25 ± 43
Vd *mL2484.43 ± 622.32 338.12 ± 167.67
F (%) *10.6
* AUC0–t (AUC0–inf), area under the analyte concentrations versus time curve from time 0 to t h (inf); MRT0–t (MRT0–inf), mean residence time at time 0–t (inf); Tmax, time of maximum concentration; T1/2, terminal half-life; Cmax, maximum concentration; CL, clearance; Vd, apparent volume of distribution; F (%), absolute bioavailability.
Table 2. Metabolite characterization of 1−(3′−bromophenyl)−heliamine by UHPLC−Q−Orbitrap−MS.
Table 2. Metabolite characterization of 1−(3′−bromophenyl)−heliamine by UHPLC−Q−Orbitrap−MS.
MouseTransformationsFragment IonError
(ppm)
Calculated
Mass (m/z)
Observed
Mass (m/z)
Formula
(M+H+)
Retention
Time
Metabolites
F *U *P *LM *
Parent333.0304, 252.1145, 179.0943, 170.9628, 164.07060.2350.0573350.0571C17H1981BrNO228.84BH
Desaturation319.0154, 286.9890, 238.0988, 170.9628, 165.0788−0.3336.0417336.0416C16H1781BrNO218.86M1−A
Desaturation319.0156, 286.9886, 238.0987, 170.9627, 165.07870.1336.0417336.0417C16H1781BrNO220.88M1−B
Desaturation, Dehydrogenation319.0022, 176.0707, 165.0789, 150.0551−0.3334.026334.0259C16H1581BrNO215.73M2−A
Desaturation, Dehydrogenation319.0024, 226.0989, 194.0727,176.0707, 161.0473,−0.7334.026334.0258C16H1581BrNO217.69M2−B
Desaturation, Dehydrogenation319.0023, 301.9997, 238.0859−0.3334.026334.0259C16H1581BrNO222.82M2−C
Desaturation, Dehydrogenation319.0024, 301.9998, 238.0858−0.3334.026334.0259C16H1581BrNO224.38M2−D
Desaturation, Desaturation,
Dehydrogenation, Dehydrogenation
236.0706, 160.0394−1.0317.9947317.9944C15H1181BrNO231.6M3
Dehydrogenation,
Dehydrogenation
329.9948, 301.99872.1346.026346.0273C17H1581BrNO232.72M4
Desaturation, Desaturation304.9995, 224.0830, 170.9627, 151.0628−0.2322.026322.0259C15H1581BrNO216.52M5
Epoxidation190.0863, 175.0628, 146.06011364.0366364.037C17H1781BrNO318.64M6
×Desaturation,
Glucuronide Conjugation
336.0454, 319.0151, 286.9885, 238.0988, 170.9627, 165.0788, −0.2512.0738512.0737C22H2581BrNO815.14M7−A
×Desaturation,
Glucuronide Conjugation
336.0416, 319.0148, 286.9885, 238.0988, 170.9627, 165.0787, −0.2512.0738512.0742C22H2581BrNO815.64M7−B
××Desaturation, Sulfation336.0416, 319.0148, 286.9890, 238.0987, 170.9627, 165.0787, 0.4415.9985415.9984C16H1781BrNO5S18.87M8−A
××Desaturation, Sulfation336.0415, 319.0161, 286.9885, 238.0988, 170.9628, 165.0788,0.1415.9985415.9991C16H1781BrNO5S20.83M8−B
×Desaturation, Dehydrogenation, Dehydrogenation, Glucuronide Conjugation332.0105, 316.9866−1.8508.0421508.0414C22H2181BrNO811.7M9−A
×Desaturation, Dehydrogenation, Dehydrogenation, Glucuronide Conjugation332.0103, 316.98671508.0421508.0437C22H2181BrNO818.61M9−B
××Desaturation, Desaturation, Glucuronide Conjugation332.0260, 304.9990, 224.0832, 170.9628, 151.0628−1.7498.0581498.0572C21H2381BrNO89.79M10−A
××Desaturation, Desaturation, Glucuronide Conjugation322.0259, 304.9997, 224.0830, 170.9627, 151.06280.2498.0581498.0582C21H2381BrNO812.11M10−B
* F, feces; U, urine; P, plasma; LM, Liver microsomes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xi, R.; Abdulla, R.; Zhang, M.; Sherzod, Z.; Ivanovna, V.V.; Habasi, M.; Liu, Y. Pharmacokinetic Study and Metabolite Identification of 1-(3′-bromophenyl)-heliamine in Rats. Pharmaceuticals 2022, 15, 1483. https://doi.org/10.3390/ph15121483

AMA Style

Xi R, Abdulla R, Zhang M, Sherzod Z, Ivanovna VV, Habasi M, Liu Y. Pharmacokinetic Study and Metabolite Identification of 1-(3′-bromophenyl)-heliamine in Rats. Pharmaceuticals. 2022; 15(12):1483. https://doi.org/10.3390/ph15121483

Chicago/Turabian Style

Xi, Ruqi, Rahima Abdulla, Miaomiao Zhang, Zhurakulov Sherzod, Vinogradova Valentina Ivanovna, Maidina Habasi, and Yongqiang Liu. 2022. "Pharmacokinetic Study and Metabolite Identification of 1-(3′-bromophenyl)-heliamine in Rats" Pharmaceuticals 15, no. 12: 1483. https://doi.org/10.3390/ph15121483

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