Научная статья на тему 'DISTRIBUTION, EXCRETION AND METABOLIC PATHWAYS OF
A SINGLE PARENTERAL ADMINISTRATION OF KAPPA-OPIOID
RECEPTOR AGONIST RU-1205'

DISTRIBUTION, EXCRETION AND METABOLIC PATHWAYS OF A SINGLE PARENTERAL ADMINISTRATION OF KAPPA-OPIOID RECEPTOR AGONIST RU-1205 Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
60
11
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
IMIDAZOBENZIMIDAZOLES / KAPPA-AGONISTS / ANALGESICS / PHARMACOKINETIC / TISSUE DISTRIBUTION / EXCRETION / BIOTRANSFORMATION

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Spasov A.A., Smirnova L.A., Grechko O.Yu., Eliseeva N.V., Lifanova Yu.V.

Introduction: The purpose was to study the pharmacokinetic properties of RU-1205 with the previously identified kappa-agonistic and analgesic effects after parenteral administration. Materials and methods: Pharmacokinetic parameters of RU-1205 after intravenous and subcutaneous administration at doses of 10 mg/kg and 50 mg/kg, respectively, were investigated, using the method of high-performance liquidchromatography with measurement of the compound according to a pre-established calibration curve. The indices of the area under the pharmacokinetic curve, clearance, half-life, residence time of the drug molecule in the body, total (apparent) volume of distribution, as well as the indicator of absolute bioavailability for subcutaneous administration were calculated. Tissue distribution and excretion of RU-1205 were also studied. Evaluation of metabolism of RU-1205 was conducted in silico, using the PALLAS 3.00 software, with the use of specific tests with CYP 450 substrates and by studying the ability of RU-1205 to form conjugates with endogenous acids. Results and discussion: It was found that after a single intravenous administration, the investigated substance was determined in the blood for 12 h; the half-life was 8.49 hours. The absolute bioavailability after subcutaneous administration is 57.35%. RU-1205 is eliminated within 3–4 days. The main route of excretion is extrarenal. The biotransformation of the substance probably proceeds mainly with the formation of oxidized forms of the initial molecule according to the reactions of the first phase of metabolic transformation, so the chance to observe phase 2 of the metabolism could be very low. Conclusion: The test substance undergoes a long process of elimination, has the highest tropism to the elimination organs and undergoes active biotransformation processes in the body of animals.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «DISTRIBUTION, EXCRETION AND METABOLIC PATHWAYS OF A SINGLE PARENTERAL ADMINISTRATION OF KAPPA-OPIOID RECEPTOR AGONIST RU-1205»

# Research Results in Pharmacology

Research Results in Pharmacology 7(2): 59-65

UDC: 615.212.7 DOI 10.3897/rrpharmacology. 7.67261

3 Research Article

Distribution, excretion and metabolic pathways of a single parenteral administration of kappa-opioid receptor agonist RU-1205

Alexander A. Spasov1, Lyudmila A. Smirnova1, Olesya Iu. Grechko1, Natalya V. Eliseeva1, Yuliya V. Lifanova1, Andrey I. Rashchenko1, Olga N. Zhukovskaya2, Anatoly S. Morkovnik2, Vera A. Anisimova2^, Olga E. Syrvacheva1

1 Volgograd State Medical University of the Ministry of Health of the Russian Federation, 1 Pavshikh Bortsov Sq., Volgograd 400131, Russia

2 Research Institute of Physical and Organic Chemistry (IPOC), Southern Federal University, 194/2 Stachki Ave., Rostov-on-Don 344090, Russia

Corresponding author: Yuliya V. Lifanova (j_semenova_pharm@mail.ru)

Academic editor: Oleg Gudyrev ♦ Received 12 March 2021 ♦ Accepted 28 May 2021 ♦ Published 29 June 2021

Citation: Spasov AA, Smirnova LA, Grechko OIu, Eliseeva NV, Lifanova YuV, Rashchenko AI, Zhukovskaya ON, Morkovnik AS, Anisimova VA, Syrvacheva OE (2021) Distribution, excretion and metabolic pathways of a single parenteral administration of kappa-opioid receptor agonist RU-1205. Research Results in Pharmacology 7(2): 59-65. https://doi.org/10.3897/rrpharmacology.7.67261

Abstract

Introduction: The purpose was to study the pharmacokinetic properties of RU-1205 with the previously identified kappa-agonistic and analgesic effects after parenteral administration.

Materials and methods: Pharmacokinetic parameters of RU-1205 after intravenous and subcutaneous administration at doses of 10 mg/kg and 50 mg/kg, respectively, were investigated, using the method of high-performance liquid chromatography with measurement of the compound according to a pre-established calibration curve. The indices of the area under the pharmacokinetic curve, clearance, half-life, residence time of the drug molecule in the body, total (apparent) volume of distribution, as well as the indicator of absolute bioavailability for subcutaneous administration were calculated. Tissue distribution and excretion of RU-1205 were also studied. Evaluation of metabolism of RU-1205 was conducted in silico, using the PALLAS 3.00 software, with the use of specific tests with CYP 450 substrates and by studying the ability of RU-1205 to form conjugates with endogenous acids.

Results and discussion: It was found that after a single intravenous administration, the investigated substance was determined in the blood for 12 h; the half-life was 8.49 hours. The absolute bioavailability after subcutaneous administration is 57.35%. RU-1205 is eliminated within 3-4 days. The main route of excretion is extrarenal. The biotransformation of the substance probably proceeds mainly with the formation of oxidized forms of the initial molecule according to the reactions of the first phase of metabolic transformation, so the chance to observe phase 2 of the metabolism could be very low.

Conclusion: The test substance undergoes a long process of elimination, has the highest tropism to the elimination organs and undergoes active biotransformation processes in the body of animals.

Keywords

imidazobenzimidazoles, kappa-agonists, analgesics, pharmacokinetic, tissue distribution, excretion, biotransformation.

Copyright Spasov AA et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

It is well know that opioid analgesics currently used in clinical practice are associated with serious nervous system related adverse events, such as euphoria, hallucinations, anxiety, respiratory depression, development of opioid addiction, abuse and misuse, and development of tolerance and physical dependence, which significantly limit the use of opioid analgesics in clinical practice. Therefore, there is an unmet medical need in development of new well-tolerated pain-relieve medications (Stein 2018).

One of the promising approaches to address these adverse events is development of new class of analgesics that are selective agonists of opioid receptors, such as kappa receptors. Unlike well-known mu- and delta- receptors agonists, these analgesics are characterised by absence of euphoria, respiratory depression and physical dependence (Prisinzano et al. 2005; Minervini et al. 2018).

RU-1205 [9-(2-morpholinoethyl)-2-(4-fluorophenyl) imidazo[1,2-a]benzimidazole] is imidazo [1,2-a] bensi-midazole based compound. In vitro, this drug was found to have a significant analgesic activity due to its agonistic effect on kappa opioid receptors (Grechko et al. 2017). In vivo, RU-1205 demonstrated a significant pain relieving effect exceeding that of butorphanol and morphine (Spasov et al. 2018), without any respiratory depression, development of analgesic tolerance and physical dependency (Grechko et al. 2017; Litvinov et al. 2017).

An analysis of pharmacokinetic parameters and tissue and organ distribution of pharmacological substance is an important and useful tool for further drug development and for a better understanding of the results of non-clinical and toxicological studies (Normative Legal Acts 2017). Understanding the pharmacokinetic characteristics can affect a decision about the way of administration of the drug and about choosing the optimal dose to be further tested in clinical trials. An analysis of tissue and organ distribution is important for a better understanding of the effects of the drug based on its mechanism of action and for predicting the potential tissue specific toxicity.

The aim of this study was to evaluate pharmacokine-tic characteristics and tissue and organ distribution of the novel therapeutic agent RU-1205 after intravenous and subcutaneous administrations.

Materials and methods

Animals

The studies were conducted on non-linear male rats, weighing 200-230 g each, received from The Laboratory Animals Nursery of the Academy of Medical and Technical Sciences (Moscow). The animals were kept at the animal testing site as per the State standard (GOST R 50258-92). All the procedures were performed after the approval by The Regional Ethic Committee (Protocol № 2077-2018 issued on 30.10.2018). The animals had constant access

to water and food and received standardized diet (granules, 5 mm in diameter, GOST R 50258-92 51899-02). The animals were deprived of food 12 hours prior to the trial.

Drugs and administration route

RU-1205 drug substance was synthesized in the Research Institute of Physical and Organic Chemistry, Southern Federal University (Fig 1). The purity was at least 99.46%.

The study drug RU-1205 was administered intravenously and subcutaneously at doses of 10 mg/kg and 50 mg/kg respectively. The drug was diluted with saline.

Figure 1. RU-1205 Structured formula. Study design

Blood and organ sampling was performed after decapitation of the animals - 5 min, 10 min, 20 min, 40 min, 1 hour, 2 hours, 4 hours, 8 hours, and 12 hours after intravenous administration, after 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, and 12 hours after subcutaneous administration and 5 minutes after administration of the saline in the control group. The rats were randomly assigned to one of the study groups (n = 5 for each time interval). 5% sodium citrate was used for blood stabilisation. Plasma was obtained by 15 min centrifugation at 3000 rpm. Drug distribution was studied in the organs responsible for excretion - liver and kidneys, in the targeted organ - brain, in the organs and tissues with variable vascularisation - heart and lungs (strong vascularisation), musculus quadriceps femoris (moderate vascularisation), and omentum (low vascularisation). The organs under study were ground and homogenized to form 20% water homogenate. The drug concentration was also evaluated in the animals' feces and urine, which were sampled in metabolic cages (Termop-last, Italy) 2, 4, 8, 12, 24, 48, 72 and 96 hours after drug administration.

The concentration of RU-1205 in plasma and 20% organ homogenate was evaluated using high performance liquid chromatography (HPLC) with a diode array UV detector (Shimadzu, Japan) on a SUPERCOSIL LC-18 column (5 ^m particle; 4.6 mm x 100 mm) at a temperature of 50 °C. For a mobile phase, acetonitrile (UF210, Russia) was used with a buffer system at a ratio of 1:1 (potassium phosphate monobasic 50 mmol, titrated with 0.5 M of NaOH solution, pH = 5.0). Before chromatography, the mobile phase was degassed by ultrasonication and filtered. The detection wavelength was set at 1 205 nm, a flow rate of 1 mL/min (Smirnova et al. 2013).

The pharmacokinetic properties of the study drug included AUC (area under the curve) (Piotrovsky 1986), Kel

(elimination constant), Cl (clearance), T1/2 (half-life of the drug), MRT (mean residence time), Vd (volume of distribution), and ft (tissue distribution).

To evaluate potential metabolites of drug RU-1205, PALLAS 3.00 software (CompuDrug Chemistry Ltd, Budapest, Hungary) was used. To evaluate the effect of RU-1205 on cytochrome P450, specific tests with CYP 450 substrates were used (Omiecinski et al. 1999; Perloff et al. 2000; Nebert et al 2002). Hexenal (MedPro Inc, Latvia) at a dose of 70 mg/kg was administered intrape-ritoneally (isoforms CYP2C9, CYP2B1 and CYP2B2). Midazolam (Roche, Switzerland) at a dose of 80 mg/kg was administered intraperitoneally (isoforms CYP3A1 and CYP2C). Alprazolam (Organica JSC, Russia) at a dose of 50 mg/kg was administered intraperitoneally (isoform CYP3A1). A possible effect of the study drug on the second biotransformation phase was evaluated by studying its ability to change the sleep onset latency and sleep duration of the UDP-glucuronosyltransferase substrate - chloral hydrate (MedPro Inc, Latvia), which had been administered intraperitoneally at a dose of 70 mg/kg.

The ability of RU-1205 to form conjugates with endogenous acids in excreta was studied using various hydrolyses of urine samples: fermentative hydrolysis, acid hydrolysis, and alkaline hydrolysis. Fermentative hydrolysis was conducted by 24-hour incubation of urine samples in the acetate buffer (0.1M, pH = 5.0) with beta-glucuronides and arylsulphatase (extracted from Helix pomatia snails, Boehringer Mannheim, Germany) at a temperature of 37 °C (Thomas et al. 1999). Acid hydrolysis was conducted by 2-hour incubation with 5M of HCl solution at a temperature of 37 °C (Dix et al. 1999; Manini et al. 1999). Alkaline hydrolysis was conducted using 5M of NaOH solution.

Statistical analyses

For the statistical analysis, the following software was used: GraphPad.Prism.5.0 and Microsoft Office Excel 2007.

Results and discussion

When evaluating the pharmacokinetic parameters of RU-1205, it was found that following intravenous administration to the study animals, it demonstrated a long plasma circulation time. A dose of 10 mg/kg of RU-1205 was traced in plasma 12 hours after administration, which confirmed a long elimination process (Fig. 2A, B). Prolonged elimination of the drug was supported by the following parameters: T1/2 and MRT were 8.49 hours and 7.14 hours, respectively (Table 1). The apparent volume of distribution (Vd=8.30 L/kg) exceeded 12-fold the real liquid volume in rats (0.67 L/kg). According to the literature, this supported that RU-1205 can penetrate intensively into organs and tissues (Davies and Morris 1993).

After subcutaneous administration of RU-1205, T1/2 was reported to be 4.28 hours and MRT - 3.1 hours. The

Figure 2. A. The mean plasma profiles of RU-1205 after intravenous (i/v) administration at a dose of 10 mg/kg. B. The mean plasma profiles of RU-1205 after subcutaneous (s/c) administration at a dose of 50 mg/kg.

Table 1. Pharmacokinetic Parameters of RU-1205 in Plasma Following Intravenous and Subcutaneous Administration in Rats

Parameter Intravenously 10 mg/kg Subcutaneously 50 mg/kg

AUC (|g*h/mL) 14.76 36.18

K, (hour-1) 0.08 0.242

Cl (L/h/kg) 0.68 2.43

T1/2 (hours) 8.49 4.28

MRT (hour) 7.14 3.1

Vd (L/kg) 8.3 13.23

F, % - 57.35

apparent volume of distribution significantly exceeded real liquid volume in the body after intravenous and subcutaneous administrations, which can indicate that RU-1205 could actively penetrate into organs and tissues. When subcutaneously administrated, the study compound is intensively absorbed from the injection site. C was

J max

reached 30 minutes after administration. Absolute bioavailability of RU-1205 was reported to be 57.35%.

When intravenously administered, the study drug distributed into all the organs and tissues. Significant amounts of the compound were detected in the targeted organ - the brain, in the elimination organs - the liver and kidneys, as well as in the omentum. RU-1205 was less absorbed by tissues with increased vascularisation (heart, lungs and spleen) and had a minimal absorption in muscle tissue.

RU-1205 was detected in the targeted organ - the brain - 5 minutes after intravenous administration of a dose of 13.9 mkg/g, and then concentration gradually decreased for 14 hours until below the level of quantitation limit (Fig. 3).

During intravenous administration, the tissue availability in the brain was 5.97 (Table 2). Cmax in the spleen, lungs and muscles was detected only within the first hour after administration of the drug. When administered intravenously, the highest concentrations of the drug was detected in the brain, kidneys and liver.

0 2 4 6 S 10 12 14 16

Time,h

Figure 3. Maximum concentration of RU-1205 in rats' brain (10 mg/kg, intravenous administration).

Table 2. RU-1205 Pharmacokinetic Parameters of Tissue Distribution After Single Intravenous Administration of 10 mg/kg

Organ AUC, ^g*h/mL Ft

Omentum 72.32 4.9

Brain 88.17 5.97

Kidneys 58.51 3.96

Liver 84.08 5.69

Heart 27.66 1.87

The results demonstrated that RU-1205 intensively distributed into organs and tissues, but its concentration in these organs was lower than in blood plasma. The compound has the lowest affinity to the heart, lungs, spleen and muscles. The highest concentration was detected in the elimination organs - the liver and kidneys. After subcutaneous administration, RU-1205 was detected in low quantities in the brain within the first three hours.

It was noted that when RU-1205 was administered in either of the administration routs mentioned above, its concentration was lower than the administered dose in blood plasma.

It was determined that RU-1205 could be detected in excrements within 72-96 hours following the administration, depending on the route of administration. It was also reported that only 0.7% of the unchanged substance was detected in urine and feces cumulatively following any of the two above routes of administration. More unchanged study drug substance was detected in urine than in excrements (Fig. 4). At the same time, it was found out that extrarenal (metabolic) clearance significantly prevailed

over renal clearance, which indicated that RU-1205 had an active metabolism (Fagerholm 2007).

Following the intravenous administration, it took a total of 3 days (72 hours) for the drug substance to be excreted, both with urine and feces (Fig. 4A). The cumulative excretion was 50.73 ^g in urine, which was approximately 2.54% of the administered dose. Hepatic clearance was 4.11 ml/h, whereas extrarenal clearance was 140.77 ml/h. The similar results were obtained after subcutaneous administration. The total excretion also took up to 72 hours (Fig. 4B). The cumulative urinary excretion was 50.63 ^g, which was 0.51% of the administered dose. Therefore, the renal clearance was only 1.399 ml/h, whereas extrarenal clearance was 274.94 ml/h.

The next part of the pharmacokinetic study was to evaluate the possible metabolism of RU-1205. The first step in silico was to conduct a computer-based prediction of potential metabolites, based on which, potential metabolites were determined. The second step involved fermentative, acid and alkaline hydrolyses of the study substance. The final step involved investigation of potential interactions of RU-1205 with specific test-substrates of some CYP450 isoforms. The substrates are known to be commonly used in other studies investigating metabolism of benzimidazole derivative compounds (Spasov et al. 2002).

We were able to detect seven possible metabolites that could be produced via an acidic reaction of hy-droxylation, using a computer-based analysis in silico. Six out of seven molecules of these metabolites were found to have chemical fragments which are likely responsible for an analgesic effect - morpholinoethyl moiety in N9 position of the molecular nucleus and fluorop-henyl moiety in the C2 atom of the condensed nucleus. During this analysis, it was also determined that during the RU-1205 metabolism, a morpholino moiety could be detached, which could lead to a decreased analgesic activity (Fig. 5).

During the second stage of the study, an ability of the drug RU-1205 to create conjugates with endogenous acids

Figure 4. Cumulative excretion of RU-1205 through kidneys and liver. A - following10 mg/kg intravenous administration; B - following 50 mg/kg subcutaneous administration. Note: GIT - gastrointestinal tract.

Table 3. Effect of CYP450 Substrates on Sleep Duration in Rats Following Administration of RU-1205

Substrate CYP 450 isoforms Control (M±m)

Changes in sleep duration Test,min Difference (M±m) (%)

Phase 1

Midazolam Alprazolam Hexenal

Phase 2

Chloral hydrate

CYP3A1, CYP2C

CYP3A1 CYP2C9, CYP2B1 and CYP2B2

Glucuronosyltrans-ferase

101.11±6.28 108.88±13.21 56.44±2.23

71.48±7.14

72.04±8.67* 175.13±8.93* 77.5±4.24*

59.59±4.45

-28.74

60.85 37.29

21.87

Figure 5. Potential metabolites of RU-1205 determined by computer modeling using Pallas 3.00.

through biotransformation was tested. Fermentative, acid and alkaline hydrolyses were conducted using rats' excreta. A chromatogram analysis, including incubation with beta-glucuronides and arylsulphatase and in combination with 5M of NaOH solution or 5M of HCl solution, demonstrated no peaks characteristic of the original chemical structure of RU-1205. Importantly, these results are similar to results obtained at the first stage (computer-based analysis), when no products of the second phase of biotransformation were detected either.

The final stage of evaluation of RU-1205 metabolism involved the investigation of the sleep duration under the influence of specific test-substrates of some of CYP450 isoforms. We determined that RU-1205 statistically significantly prolonged the sedative effect of hexenal (CYP2C9, CYP2B1, and CYP2B2) and alprazolam (CY-P3A1) by 37.29% and 60.85%, respectively. However, in the combination with midazolam (CYP3A1 and CYP 2C), the sleep duration reduced by 28.74% (Table 3).

When RU-1205 was administered in combination with specific test-substrates of CYP450 isoforms, we found out that the drugs were likely metabolised though P450 CYP3A1 cytochrome isozyme (xenobiotic oxidation). CYP2B cytochromes, responsible for hydroxylation of xenobiotics, were less likely to participate in RU-1205 metabolism. Sleep duration following the midazolam and RU-1205 administration did not change significantly, which supported the hypotheses that CYP2C cytochro-mes are unlikely to participate in RU-1205 metabolism (xenobiotic oxidation). Changes in sleep duration following the administration of chloral hydrate and RU-1205 were not statistically significant either, which supported

Note: * - statistically significant when compared with control, U Mann-Whitney test (p<0.05).

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

the hypothesis that RU-1205 was unlikely to produce glu-curonyl-conjugated metabolites and, therefore, the chance to observe the phase 2 reactions in the metabolism of the study substance was very low.

It was concluded that based on the results of the first phase of metabolic transformation involving CYP3A1 cytochrome, RU-1205 biotransformation is likely to involve oxygenation forms of the original formula. RU-1205 is unlikely to undergo synthetic reactions during the second phase of biotransformation, nor are new conjugates produced.

Conclusion

Therefore, this study involved the evaluation of the pharmacokinetic characteristics of new drug RU-1205, with its singular intravenous and subcutaneous administrations. Tissue and organ distribution of the study substance in rats and elimination of the drug were examined. Total bioavailability following subcutaneous administration was determined, and the potential metabolic pathways were predicted.

Based on the study results, some recommendation for using RU-1205 as an injection in clinical setting can be given. The sesults of this study also can be used for further prediction of potential interactions of RU-1205 via CYP3A1, CYP2B1, and CYP2B2 cytochromes. These results can play a significant role in planning first-in-human clinical trials.

Acknowledgments

RU 1205 was synthesized with financial support by the Ministry of Science and Higher Education of the Russian Federation, grant FENW-2020-0031 (0852-2020-0031).

Conflict of interests

The authors declare no conflict of interests.

References

■ Grechko OY, Litvinov RA, Spasov AA, Rashchenko AI, Shtareva DM, Anisimova VA (2017) Study of and S-Opioid activities in agents with various K-receptor selectivity. Bulletin of Experimental Biology and Medicine 162: 632-635. https://doi.org/10.1007/ s10517-017-3674-5 [PubMed]

■ Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharmaceutical Research 10(7): 1093-1095. https://doi.org/10.1023/A:1018943613122 [PubMed]

■ Dix KJ, Coleman DP, Jeffcoat AR (1999) Comparative metabolism and disposition of gemfibrozil in male and female Sprague-Dawley rats and Syrian Golden hamsters. Drug Metabolism and Disposition 27(1): 138-146. [PubMed]

■ Fagerholm U (2007) Prediction of human pharmacokinetics - renal metabolic and excretion clearance. The Journal of Pharmacy and Pharmacology 59(11): 1463-1471. https://doi.org/10.1211/ jpp.59.11.0002 [PubMed]

■ Litvinov RA, Eliseeva NV, Grechko OYu, Spasov AA (2017) Influence of the kappa-opioid agonist RU-1205 compound in wide spread of doses on the effects of neuromediator analyzers: Bechavioral testing. Clinical Research Trials 3(6): 1-7. https://doi.org/10.15761/ CRT. 1000202

■ Manini P, Andreoli R, Mutti A, Bergamaschi E, Franchini I (1999) Determination of free and glucuronated hexane metabolites without prior hydrolysis by liquid- and gas-chromatography coupled with mass spectrometry. Toxicology Letters 108(2-3): 225-231. https:// doi.org/10.1016/S0378-4274(99)00093-4 [PubMed]

■ Minervini V, Lu HY, Padarti J, Osteicoechea DC, France CP (2018) Interactions between kappa and mu opioid receptor agonists: effects of the ratio of drugs in mixtures Psychopharmacology (Berl) 235(8): 22452256. https://doi.org/10.1007/s00213-018-4920-x [PubMed] [PMC]

■ Nebert DW, Russell DW (2002) Clinical importance of the cytochromes P450. Lancet 360(9340): 1155-1162. https://doi. org/10.1016/S0140-6736(02)11203-7 [PubMed]

■ Normative Legal Acts in the Field of Drug Circulation Within the Framework of the Eurasian Economic Union (2017) Volume 2. Development and Research of Medicinal Products. Remedium, Moscow, 308 pp. [in Russian]

Author contributions

■ Everyone participated in the discussion of the results.

■ Omiecinski CJ, Remmel RP, Hosagrahara VP (1999) Concise review of the cytochrome P450s and their roles in toxicology. Toxicologi-cal Sciences 48(2): 151-156. ttps://doi.org/10.1093/toxsci/48.2.151 [PubMed]

■ Perloff MD, Moltke LL, Court MH, Kotegawa T, Shader R, Green-blatt DJ (2000) Midazolam and triazolam biotransformation in mouse and human liver microsomes: relative contribution of CYP3A and CYP2C isoforms. The Journal of Pharmacology and Experimental Therapeutics 292(2): 618-628. [PubMed]

■ Piotrovsky VK (1986) Method of statistical moments and integral model-independent parameters of pharmacokinetics. Pharmacology and Toxicology [Farmakologiya i Toksikologiya] 49(5): 118-127. [in Russian]

■ Prisinzano TE, Tidgewell K, Harding WW (2005) Opioids as potential treatments for stimulant dependence. The AAPS Journal 7(3): 592-599. https://doi.org/10.1208/aapsj070361 [PubMed]

■ Smirnova LA, Spasov AA, Rashchenko AI, Suchkov EA, Ryabukha AF, Kuznetsov KA (2013) Analytical features of the quantitative determination of benzimidazole derivatives by high performance liquid chromatography. Volgograd Journal of Medical Scientific Research [Volgogradskii Nauchno-Meditsinskii Zhurnal] 2(38): 9-13. [in Russian]

■ Spasov AA, Grechko OYu, Shtareva DM, Rashchenko AI, Eliseeva NV, Anisimova VA (2018) Analgesic activity of the kappa opioid receptor agonist - RU-1205 in rats. Journal of Clinical and Health Sciences 3(2): 13-18. https://doi.org/10.24191/jchs.v3i2.7275

■ Spasov AA, Smirnova LA, Iyozhytsa IN, Sergeeva SA, Ozerov AA (2002) Pharmacokinetics of some benzimidazole derivatives. Questions of Medicinal Chemistry [Voprosy Meditsinskoi Khimii] 48(3): 233-258. [in Russian]

■ Stein C (2018) New concepts in opioid analgesia. Expert Opinion on Investigational Drugs 27(10): 765-775. https://doi.org/10.1080/135 43784.2018.1516204 [PubMed]

■ Thomas BF, Burgess JP, Coleman DP, Scheffler NM, Jeffcoat AR, Dix KJ (1999) Isolation and identification of novel metabolites of gemfibrozil in rat urine. Drug Metabolism and Disposition 27(1): 147-157. [PubMed]

■ Alexander A. Spasov, Doctor Habil. of Medical Sciences, Full Professor, Academician of the Russian Academy of Sciences, Head of the Department of Pharmacology and Bioinformatics, e-mail: aspasov@mail.ru, ORCID ID http://orcid.org/0000-0002-7185-4826. Concept and design of the manuscript, approval of the final version.

■ Lyudmila A. Smirnova, Doctor Habil. of Biological Sciences, Head of the Laboratory of Pharmacological Kinetics, Research Institute of Pharmacology, e-mail: smirlusi@yandex.ru. Concept and manuscript design.

■ Olesya Iu. Grechko, Doctor Habil. of Medical Sciences, Full Professor of the Department of Pharmacology and Bioinformatics, e-mail: olesiagrechko@mail.ru, ORCID ID https://orcid.org/0000-0002-4184-4897. Draft ofthe manuscript.

■ Natalya V. Eliseeva, PhD in Medical Sciences, Associate Professor of the Department of Pharmacology and Bioinformatics, e-mail: nvkirillova@rambler.ru, ORCID ID https://orcid.org/0000-0002-2243-5326. Obtaining, analyzing and interpreting data, revising the manuscript

■ Yuliya V. Lifanova, PhD student in Medical Sciences, Assistant Professor of the Department of Pharmacology and Bioinformatics, e-mail: j_semenova_pharm@mail.ru, ORCID ID https://orcid.org/0000-0001-9663-5067. Obtaining data and revising the manuscript.

■ Andrey I. Rashchenko, PhD in Pharmacy, Senior Researcher, Laboratory of Pharmacology of Antioxidant Agents, Research Institute of Pharmacology, e-mail: a.rashencko@yandex.ru, ORCID ID https://orcid.org/0000-0001-8357-4007. Conducting an experimental series for the study of pharmacokinetic parameters and processing the results obtained.

■ Olga N. Zhukovskaya, PhD in Chemical Sciences, Researcher in the Laboratory of Organic Synthesis, e-mail: zhukowskaia.ol@yandex.ru, ORCID ID http://orcid.org/0000-0003-0865-6656. The author took part in synthesis of the substances.

■ Anatoly S. Morkovnik, Doctor Habil. of Chemical Sciences, Head of the Laboratory of Organic Synthesis, e-mail: asmorkovnik@sfedu.ru, ORCID ID https://orcid.org/0000-0002-9182-6101. Design of molecule and direct synthesis.

■ Vera A. Anisimova, PhD in Chemical Sciences, ORCID ID https://orcid.org/0000- 0002-0877-4575. Design of molecule and direct synthesis.

■ Olga E. Syrvacheva, co-investigator at the Department of Pharmacology and Bioinformatics, e-mail: xelga4e@ yandex.ru, ORCID ID https://orcid.org/0000-0001-6808-5504. Obtaining data and revising the manuscript.

i Надоели баннеры? Вы всегда можете отключить рекламу.