Next Article in Journal
Novel Anthranilamide-Based FXa Inhibitors: Drug Design, Synthesis and Biological Evaluation
Previous Article in Journal
Microwave-Assisted Synthesis of Bioactive Six-Membered Heterocycles and Their Fused Analogues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis and Biological Evaluation of Brain-Targeted Thiamine Disulfide Prodrugs of Ampakine Compound LCX001

Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
*
Author to whom correspondence should be addressed.
Molecules 2016, 21(4), 488; https://doi.org/10.3390/molecules21040488
Submission received: 26 February 2016 / Revised: 30 March 2016 / Accepted: 8 April 2016 / Published: 14 April 2016
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Ampakine compounds have been shown to reverse opiate-induced respiratory depression by activation of amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors. However, their pharmacological exploitations are hindered by low blood-brain barrier (BBB) permeability and limited brain distribution. Here, we explored whether thiamine disulfide prodrugs with the ability of “lock-in” can be used to solve these problems. A series of thiamine disulfide prodrugs 7a7f of ampakine compound LCX001 was synthesized and evaluated. The trials in vitro showed that prodrugs 7e, 7d, 7f possessed a certain stability in plasma and quickly decomposed in brain homogenate by the disulfide reductase. In vivo, prodrug 7e decreased the peripheral distribution of LCX001 and significantly increased brain distribution of LCX001 after i.v. administration. This compound showed 2.23- and 3.29-fold greater increases in the AUC0-t and MRT0-t of LCX001 in brain, respectively, than did LCX001 itself. A preliminary pharmacodynamic study indicated that the required molar dose of prodrug 7e was only one eighth that of LCX001 required to achieve the same effect in mice. These findings provide an important reference to evaluate the clinical outlook of ampakine compounds.

Graphical Abstract

1. Introduction

The use of opioid analgesics is still the most effective way to manage moderate to severe pain in the clinic [1]. Nevertheless, opiate-induced respiratory depression is a life-threatening condition and a leading cause of death that may arise not only from overdoses, but also during routine procedures supervised by clinicians, including surgical anaesthesia, post-operative analgesia, and as a result of routine out-patient management of pain from cancer, accidents, or other illnesses [2,3,4,5]. Naloxone and related opioid antagonists are currently used to counter respiratory depression in an emergency setting [6]. However, those agents reverse analgesia, so selective antagonism of the respiratory depressive effects of opioids without decreasing their analgesic effects would be a major step toward improving opioid safety.
Recent studies demonstrated that AMPA receptor was a critical component for the generation of respiratory rhythm within the pre-Botzinger complex [7,8,9,10], and was becoming an ideal target for reversing respiratory depression. Ampakine compounds, developed by Cortex Pharmaceuticals, such as CX717, CX1739 and CX1942 etc., had been proved to modulate respiratory drive and rhythmogenesis positively by activation of AMPA receptor [11,12,13,14,15,16,17,18]. Most advanced ampakine compounds are CX717 and CX1739 [19], both of which are currently in Phase II clinical trials. Studies demonstrated that a single oral dose of 1500 mg of CX717 had positive effects on respiratory depression induced by pain relieving opiates, without affecting the pain relieving effects [14]. The reversal degree of the basal respiratory rate was similar to that obtained with the opioid antagonist, naloxone. Similar studies in respiratory depression with CX1739 at a high dose of 900 mg have also been planned [20].
Structurally these compounds belong to the benzamide derivative type 1 (Figure 1), but their specific structures remained unknown. According to Cortex’s patents [21,22], we found that compound 2 (Figure 1, LCX001) was one of the most active compounds, with the best electrophysiological effect giving a 21% increase in amplitude of the field excitatory postsynaptic potential (EPSP) in the rat dentate gyrus and producing a 100% inhibition of damphetamine- stimulated locomotion in the behavioral testing [21], so we infered that LCX001 was probably an analogue of the clinical compounds or even one of them. To date, no further developments for these compounds were reported for this indication. We assumed that besides poor absorption the low blood-brain barrier permeability (tPSA of LCX001: 74.49) and limited brain distribution might hinder the development of the compounds. As a result, high-dose use of drugs and non-targeting distribution could cause serious safety problems. In 2007, Food and Drug Administration (FDA) even rejected an investigational new drug (IND) application for CX717 as a treatment for attention-deficit hyperactivity disorder (ADHD) because of its toxicity after chronic dosing at very high dose levels in animals.
The thiamine disulfide system (TDS), has been widely applied as a classic brain-targeted prodrug strategy to a variety of drugs such as DOPA, naproxen, ibuprofen (Figure 1) etc. [23,24,25]. In this approach, the increased lipophilic nature of the prodrugs could facilitate passive diffusion across the BBB; subsequently, TDS was reduced and ring-closed to be a thiazolium by disulfide reductase once across the BBB, and then the prodrugs with this system were “locked” in the brain where they can provide sustained release of the active drug via hydrolysis (Figure 2) [23].
In this paper, we introduced the lipophilic TDS with “lock-in” ability to mediate the penetration of LCX001 and designed some brain-targeted prodrugs 7a7f (Figure 1), which were expected to have synergism and attenuation effects and also provide an important reference value to evaluate the clinical outlook of ampakine compounds. These designed compounds were sequentially synthesized. The in vitro stabilities of these prodrugs were discussed. Furthermore, this account also included the biodistribution and pharmacodynamic study of these prodrugs in mice after i.v. administration.

2. Results and Discussion

2.1. Chemistry

The synthetic routes for the prodrugs 7a7f are shown in Scheme 1. As shown, N,4-dimethyl-5-(2-(hydroxy)ethyl)thiazolium iodide (9) can be obtained through methylation of 5-(2-hydroxyethyl)-4-methylthiazole (8). Halohydrocarbons 10a10f and sodium thiosulfate pentahydrate were mixed and refluxed for 7 h to afford intermediates 11a11f. Thiamine disulfide derivatives 12a12f were obtained by the reaction of N,4-dimethyl-5-(2-(hydroxy)ethyl) thiazolium iodide (9) with intermediates 11a11f in the presence of NaOH at room temperature. Thiamine disulfide derivatives 12a12f further react with glutaric anhydride to give the requisite intermediate 13a13f. These compounds were conjugated with LCX001 in the presence of EDCI, HOBT, DMAP and triethylamine to afford the prodrugs 7a7f. The structures of all the synthesized prodrugs 7a7f have been confirmed by NMR, and MS, and all the corresponding data can be found in the Experimental Section.

2.2. Metabolism Studies of the Prodrugs

2.2.1. Metabolic Stability

To determine the plasma stability and brain stability of these prodrugs, we assayed the pharmacokinetics of the prepared compounds 7a7f in mice plasma extract and brain homogenate at 37 °C. The pseudo-first order rate constants of disappearance (Ke) of the prodrugs and their half lives (t1/2) which were calculated by linear regression of Ln of peak area against time in minutes are listed in Table 1.
In plasma, compounds 7a7f decomposed at a high rate. This instability is due to the existence of ester and disulfide linkages in 7a7f. The stability of these prodrugs had a descending trend of 7d (t1/2 = 15.41 min) > 7c (t1/2 = 10.24 min) > 7b (t1/2 = 9.82 min) > 7a (t1/2 = 2.23 min), indicating that the longer the carbon chain, the better the plasma stability. Larger steric hinderance produced by longer carbon chain hindered the reduction of the disulfide bond. The half-lives of 7d and 7e were similar, suggesting that substitution in the side chain had little influence on the plasma stability. We tried to further increase the plasma stability of the compounds by introducing the benzyl group, but no effect was obvious. In spite of a relatively high metabolic rate, it is reasonable to give enough time (t1/2 = 15.41 min–15.97 min) for the compounds 7d, 7e, 7f to be distributed and reach the brain before complete decomposition.
In brain homogenate, all the compounds decomposed swiftly (t1/2 = 3.48 min–18.78 min) probably due to the abundance of disulfide reductase in brain. They underwent the expected reduction and cyclization of the thiamine disulfide component to the corresponding thiazolium in brain homogenate. These compounds could be reduced to strongly polar products with high rates, and thus, contributed to the “lock-in” effect in the brain before they were effluxed to the periphery.

2.2.2. Release of the Parent Drug by Prodrugs in Brain Homogenate

To study the release of LCX001 by the prodrugs in brain, the peak in the LC-MS/MS corresponding to the parent compound was also analyzed. A certain increase in peak area over time was observed, indicating that the prodrugs were indeed converted to the parent drug by esterases in mice brain homogenate. Although the deliverance of LCX001 seemed not fast, the concentration increased steadily as time went by, which might indicate sustained release of LCX001 by the prodrugs in the brain. The release rates of different produgs were similar, which indicated that the release process of parent drug was unlikely to be seriously affected by the formation of a thiazolium cation. Peak integrals were normalized by dividing by the area for the peak of each prodrug itself at 5 min. The ratios are shown in Figure 3.
Several conclusions could be drawn from the above results: first, the half-lives of the prodrugs were mainly influenced by the length of the carbon chains linked to the disulfide functional group. Second, disulfide reductase produced reduction and ring closure of compounds 7a7f predominated over their clearance in brain with high efficiency, since the decomposition of the prodrugs consisted of reduction and hydrolysis, and the hydrolysis occurs at a relatively low rate. Finally, the release process of the parent drug was unlikely to be seriously affected by the formation of a thiazolium ion. As the above in vitro studies demonstrated that prodrugs 7e, 7d, 7f possess preliminary and favorable physicochemical properties, we selected compound 7e as a model compound to proceed with an in vivo study.

2.3. In Vivo Studies

2.3.1. Pharmacokinetics in Plasma of the Prodrug 7e

To understand the in vivo behavior of the prodrug, we assessed the plasma pharmacokinetics of LCX001 and prodrug 7e in mice (Figure 4).
The compounds were injected through the caudal vein of the mice with a single dose equivalent to 1 mg/kg body weight of LCX001, and then blood was collected to analyze the concentration of LCX001 at different intervals by an LC-MS/MS method. The pharmacokinetic parameters of LCX001 in blood are reported in Table 2.
Compared with the concentration curve of LCX001 after administration of underivatized LCX001, prodrug 7e exhibited significantly lower concentrations at different time intervals. Free LCX001 of prodrug 7e and LCX001 presented with an area under the concentration-time profile (AUC0-t) ratio of 0.40 and the mean residence times (MRT) of LCX001 in plasma after i.v. administration of 7e was 0.77 times that of LCX001, indicating that 7e could increase brain distribution because of its increased lipophilicity and be cleared more quickly than LCX001 due to the strongly polar thiazolium cation produced by reduction of the corresponding thiamine disulfides.

2.3.2. Brain Distribution of Prodrug 7e

To further evaluate the possibility of prodrug 7e being transported across the BBB, the brain distribution of prodrug 7e and LCX001 after injection were detected at fixed time intervals. The concentrations of LCX001 in brain versus time curves are displayed in Figure 5. The curves show that prodrug 7e exhibited much higher distribution in brain. The pharmacokinetic parameters of LCX001 in brain are reported in Table 3. The data showed that Cmax of LCX001 in brain after i.v. administration of prodrug 7e was not significantly improved, but the AUC0-t and MRT0-t of LCX001 was significantly higher than that after the injection of the parent compound LCX001. The AUC0-t for prodrug 7e was enhanced to 2.23 times that of LCX001. The MRT0-t was increased to 3.29 times that of LCX001. The curves displayed that LCX001 could be quickly metabolized or effluxed while prodrug 7e showed a certain stability. This agreed well with the fact that the “lock-in” effect and sustained release effect of the prodrug 7e could effectively delay the clearance of LCX001.

2.4. Pharmacodynamic Study of Prodrug 7e

To investigate the effect of 7e on opiate-induced respiratory depression, we proceeded with a preliminary pharmacodynamic study in mice. The potent opiate receptor agonist 030418 (Ki = 0.91 nM for μ-opioid receptor) [26] was administered to induce fatal apneas. Varying doses of prodrug 7e were administered 10 min before 030418 administration. The effect of prodrug 7e and LCX001 on opiate-induced respiratory depression was evaluted by the mortality of mice. The results are reported in Table 4. The data suggested that prodrug 7e reduced the death rate in a dose-dependent manner. Moreover, 3 mg/kg prodrug 7e gave the same effect as 10 mg/kg LCX001 on reducing mortality, which indicated that the required molar dose of 7e was only one eighth of that of LCX001 to achieve the same effect. Further pharmacodynamic studies are currently underway.

3. Experimental Section

3.1. General Information

TLC was performed using precoated silica gel plates (Yantai dexin Bio-Technology Co., Ltd., Yantai, China). Column chromatography was performed using silica gel (200–300 mesh; Yantai Chemical Industry Research Institute, Yantai, China). Melting points were measured on a RY-1 apparatus (Tianda Tianfa Technology Co., Ltd., Tianjin, China). NMR spectra were recorded on a JNM-ECA-400 400 MHz spectrometer (JEOL Ltd., Tokyo, Japan) using CDCl3, DMSO-d6 and D2O as solvent. Chemical shifts are expressed in δ (ppm), with tetramethylsilane (TMS) functioning as the internal reference, coupling constants (J) were expressed in Hz. Mass spectroscopy (MS) was carried out on an API 3000 triple-quadrupole mass spectrometer (AB Sciex, Concord, ON, Canada) equipped with a Turbo IonSpray electrospray ionization (ESI) source was used for mass analysis and detection. Analyst 1.4 software (AB Sciex) was used for data acquisition.

3.2. Liquid Chromatographic Conditions

An Agilent 1100 system (Agilent Technologies, Palo Alto, CA, USA) consisting of a vacuum degasser, a quaternary pump, and an autosampler was used for HPLC solvent and sample delivery. Chromatographic separation was achieved using a Poroshell 120 C18 column (50 mm × 2.1 mm, 2.7 μm, Agilent) and a C18 guard column (4 × 3.0 mm, 5 μm; Phenomenex, Torrance, CA, USA). The column temperature was maintained at 25 °C. Gradient elution was performed using purified water as solvent A and acetonitrile as solvent B. Two elution gradient programs were used. The program applied for in vitro samples was as follows: 0.0–0.5 min 10% B to 80% B; 0.5–1.5 min 80% B; 1.5–1.6 min 80% B to 10% B; 1.6–6.0 min 10% B. And the program applied for in vivo samples was as follows: 0.0–0.5 min 10% B to 80% B; 0.5–1.3 min 80% B; 1.3–1.5 min 80% B to 10% B; 1.5–7.0 min 10% B. The flow rate was set at 0.3 mL/min. The injection volume was 10 μL.

3.3. Mass Spectrometric Conditions

The mass spectrometer was operated in the positive-ion mode. Ultrapure nitrogen was used as a nebulizer, curtain, and collision-activated dissociation (CAD) gas at 8, 10, and 6 instrument units, respectively. The optimized Turbo IonSpray voltage and temperature were set to 4500 V and 450 °C, respectively. Quantification was performed with multiple reaction monitoring. Glipizide was chosen as internal standard. The transitions and the optimized collision energies (CE) for each compound were listed in Table 5. Each transition was monitored with a 150-ms dwell-time.

3.4. Stability in Plasma Extract and Brain Homogenate

Blood was drawn from mice though the orbital sinus and was collected in a heparinized tube paved with heparin sodium. Samples were centrifuged at 10,000 rpm for 16 min to separate plasma which was diluted with four volumes of water. The brain was removed and homogenized. Samples were then placed on ice and used immediately. Ten μL acetonitrile solution (10 μg/mL) of the compound was added to 50 μL of plasma or brain homogenate and gently vortexed. Samples were incubated at 37 °C and aliquots were removed after 0, 5, 15, 30, 60 and 90 min, respectively. One hundred μL acetonitrile was added to each and vortexed. Samples were centrifuged for 10 min to remove proteins and the supernatants were analyzed by LC-MS/MS method as described.

3.5. Release of the Parent Drug by Prodrugs in Brain Homogenate

The prodrugs were treated in brain homogenate according to the procedure in Section 3.4 except that the intervals were changed to 0, 5, 15, 30 and 60 min. The peak of LCX001 was analyzed by LC-MS/MS described in Section 3.2 and Section 3.3. Peak integrals were normalized by dividing by the area for the peak of each prodrug itself at 5 min.

3.6. Biodistribution Studies in Vivo

3.6.1. Test Animals

Adult Kunming mice weighing 20–22 g were obtained from the animal center of academy of military medical sciences. The animals were left for two days to acclimatize to animal room conditions and were maintained on standard pellet diet and water ad libitum. Food was withdrawn on the day before the experiment, but free access to water was allowed. Since the experiments could be completed within 24 h, there was no significant change in the mice body weight during the experiment. All animals received human care, and the study protocols complied with the guidelines of the animal center of academy of military medical sciences. Throughout the experiments, the animals were handled according to the international ethical guidelines for the care of laboratory animals.

3.6.2. Biodistribution Studies of the Prodrug and LCX001

Mice were randomly divided into two groups, 27 in each group, for different sampling times and housed in one cage. Each animal was injected with 7e or LCX001 in 20% w/v hydroxypropyl-β-cyclodextrin and saline (0.45%) solution through the tail vein at a single dose equivalent to 1 mg/kg body weight of LCX001. At appropriate time intervals (5, 10, 30, 45, 60, 90, 120, 240 and 480 min), the animals were sacrificed and 1 mL blood samples withdrawn from the orbital sinus were collected in heparinized tubes. Fifty μL plasma was immediately separated by centrifugation, diluted with 200 μL methanol and stored at −20 °C until assay. Meanwhile, the brain was removed, weighed, homogenized and diluted with methanol to 1:5 (g/mL). The homogenates were also stored at −20 °C until assay. Internal standard (200 ng/mL) was added to the samples. The samples were vortexed for 3 min and then centrifuged at 9500 rpm for 15 min. Fifty μL supernatants were withdrawn, then 50 μL of water was added to each aliquot and vortexed. Ten μL samples were analyzed by LC-MS/MS method as described.

3.7. Statistical Analysis

The Cmax, AUC0-t, Tmax, MRT0-t and t1/2 were calculated by WinNonlin software (version 5.2.1, Pharsight, Mountain View, CA, USA). Statistical evaluation was performed using analysis of variance followed by t-test. A value of p < 0.05 was considered significant.

3.8. Pharmacodynamic Study of Prodrug 7e

3.8.1. Test Animals

A total of 80 male Kunming mice weighing 20–22 g were used in this study. Animals were fed ad libitum with a commercial rodent feed and had free access to drinking water. All animals received human care, and the study protocols complied with the guidelines of the animal center of academy of military medical sciences. Throughout the experiments, the animals were handled according to the international ethical guidelines for the care of laboratory animals.

3.8.2. Model of Opiate-Induced Respiratory Depression

Respiratory depression in conscious mice was induced by hypodermic (15 mg/kg) administration of 030418. Ten min prior to 030418 administration, animals were randomly assigned to the experimental groups and received an intravenous injection of either saline solution (control), LCX001 (10 mg/kg), prodrug 7e (30 mg/kg, 10 mg/kg, 3 mg/kg, 1 mg/kg or 0.5 mg/kg) (Table 4).

3.8.3. Assessment of the Prodrug 7e Effect

During the experiment, the poisoning time, poisoning symptoms and death time were recorded. The death rate (death number/sample number) was used to evaluate the effect of 7e on opiate-induced respiratory depression.

3.9. Synthesis

N,4-Dimethyl-5-(2-(hydroxy)ethyl)thiazolium iodide (9). 5-(2-Hydroxyethyl)-4-methylthiazole (100.0 g, 698.2 mmol) and methyl iodide (100.0 mL, 1500.0 mmol) were mixed and refluxed for 2 h. After evaporation of excess methyl iodide, to the residual brown syrup was added ether (100 mL) and stirred for 30 min, the precipitate was filtered to yield 9 as a pale-yellow solid (191.4 g, 96.2%). Mp: 82–84 °C. 1H-NMR (DMSO-d6, ppm): 2.43 (s, 3H), 3.03 (t, 2H, J = 5.5 Hz), 3.63 (t, 2H, J = 5.6 Hz), 4.09 (s, 3H), 9.96 (s, 1H); MS m/z [M]+ calculated for C7H12NS+: 158.1; found: 158.1.
General procedure for synthesis of S-substituted sodium thiosulfates 11a11f. A solution of sodium thiosulfate pentahydrate (24.8 g, 100.0 mmol) in water (60 mL) was added to the solution of alkyl bromide (100 mmol) in ethanol (30 mL) under stirring. The reation mixture was heated to 110 °C and maintained for 7 h. After evaporation of the solvent, the residual white solid was dried in a vacuum desiccator to yield S-substituted sodium thiosulfates 11a11f.
General procedure for synthesis of compounds 12a12f. Under argon protection, to a 30 mL aqueous of quaternary ammonium salt 9 (14.3 g, 50.0 mmol) and sodium hydroxide (4.0 g, 100.0 mmol) was added S-substituted sodium thiosulfates 11a11f (120.0 mmol). The reaction mixture was stirred for 1 h at room temperature. The resultant oily substance was extracted with ethyl acetate (450 mL) and the organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure. Purification of the crude product by column chromatography with dichloromethane-methanol (100:1) as eluent gave a yellow oil.
N-(3-(Ethyldisulfanyl)-5-hydroxypent-2-en-2-yl)-N-methylformamide (12a, Figure S1). 40.9% yield as a yellow oil. 1H-NMR (CDCl3, ppm): 1.19 (t, 3H, J = 7.3 Hz), 1.96, 1.86 (2s, 3H), 2.64 (q, 2H, J = 7.3 Hz), 2.70 (t, 2H, J = 6.7 Hz), 2.98, 2.83 (2s, 3H), 3.55–3.43 (m, 2H), 7.96, 7.84 (2s, 1H). MS m/z [M + H]+ calculated for C9H17NO2S2: 236.1; found: 236.2.
N-(3-(Propyldisulfanyl)-5-hydroxypent-2-en-2-yl)-N-methylformamide (12b, Figure S2). 64.8% yield as a yellow oil. 1H-NMR (CDCl3, ppm): 0.97 (t, 3H, J = 7.3 Hz), 1.69–1.63 (m, 2H), 2.01, 1.96 (2s, 3H), 2.47 (br, 1H), 2.59 (t, 2H, J = 7.6 Hz), 2.87 (t, 2H, J = 6.7 Hz), 3.06, 2.95 (2s, 3H), 3.81 (t, 3H, J = 6.4 Hz), 7.96, 7.92 (2s, 1H); MS m/z [M + H]+ calculated for C10H19NO2S2: 250.1; found: 250.1.
N-(3-(Butyldisulfanyl)-5-hydroxypent-2-en-2-yl)-N-methylformamide (12c, Figure S3). 70.5% yield as a yellow oil. 1H-NMR (CDCl3, ppm): 0.91(t, 3H, J = 7.3 Hz), 1.43–1.33 (m, 2H), 1.65–1.57 (m, 2H), 2.01, 1.96 (2s, 3H), 2.51 (br, 1H), 2.62 (t, 2H, J = 7.3 Hz), 2.87 (t, 2H, J = 6.7 Hz), 3.06, 2.95 (2s, 3H), 3.81 (t, 2H, J = 6.8 Hz), 7.96, 7.92 (2s, 1H). MS m/z [M + H]+ calculated for C11H21NO2S2: 264.1; found: 264.2.
N-(3-(Amyldisulfanyl)-5-hydroxypent-2-en-2-yl)-N-methylformamide (12d, Figure S4). 52.0% yield as a yellow oil. 1H-NMR (CDCl3, ppm): 0.90 (t, 3H, J = 7.3 Hz), 1.36–1.29 (m, 4H), 1.65–1.60 (m, 2H), 2.01, 1.97 (2s, 3H), 2.49 (br, 1H), 2.61 (t, 2H, J = 7.3 Hz), 2.87 (t, 2H, J = 6.7 Hz), 3.06, 2.95 (2s, 3H), 3.80 (t, 2H, J = 6.8 Hz), 7.96, 7.92 (2s, 1H). MS m/z [M + H]+ calculated for C12H23NO2S2: 278.1; found: 278.0.
N-(3-Isoamyldisulfanyl)-5-hydroxypent-2-en-2-yl)-N-methylformamide (12e, Figure S5). 76.8% yield as a yellow oil. 1H-NMR (CDCl3, ppm): 1.19 (t, 3H, J = 7.3 Hz), 1.53–1.47 (m, 2H), 1.68–1.61 (m, 1H), 2.01, 1.96 (2s, 3H), 2.49 (br, 1H), 2.62 (t, 2H, J = 7.6 Hz), 2.88 (t, 2H, J = 6.8 Hz), 3.06, 2.95 (2s, 3H), 3.80(t, 3H, J = 6.4 Hz), 7.96, 7.92(2s, 3H). MS m/z [M + H]+ calculated for C12H23NO2S2: 278.1; found: 278.0.
N-(3-(Benzyldisulfanyl)-5-hydroxypent-2-en-2-yl)-N-methylformamide (12f, Figure S6). 44.1% yield as a yellow oil. 1H-NMR (d6-DMSO, ppm): 1.91, 1.85 (2s, 3H), 2.62 (t, 2H, J = 6.7 Hz), 2.82, 2.81 (2s, 3H), 3.52–3.47 (m, 2H), 3.92 (s, 2H), 4.74–4.70 (m, 1H), 7.35–7.25 (m, 5H), 7.96, 7.81 (2s, 1H). MS m/z [M + H]+ calculated for C14H19NO2S2: 298.1; found: 298.2.
General procedure for synthesis of 4-(N-methylformamido)pent-3-en-1-yl)oxy)-5-oxopentanoic acids 13a13f. A stirred solution of intermediate 12a12f (10.0 mmol) in dry dichloromethane (50 mL) was treated with DMAP (0.1 g, 0.8 mmol) and glutaric anhydride (3.4 g, 30.0 mmol). The mixture was refluxed for 5 h. The solvent was evaporated under vacuum and purification of the crude product by column chromatography with dichloromethane-methanol (100:1) as eluent gave 13a13f as yellow syrup.
General procedure for synthesis of compounds 7a7f. The intermediate 13a–13f (4.7 mmol), LCX001 (1.0 g, 3.6 mmol), triethylamine (0.5 mL, 3.6 mmol) and HOBT (486 mg, 3.6 mmol) were dissolved in dry dichloromethane (50 mL) in a round-bottomed flask under string for 10 min. After addition of DMAP (44 mg, 0.36 mmol) and EDCI (1.1 g, 5.4 mmol), the mixture was refluxed for 5 h. The resulting mixture was concentrated by a rotary evaporator to afford yellow oleosus residue that was purified by column chromatography on silica gel (ethyl acetate/petroleum ether, (1:1)) to provide 7a7f as white solids.
S-3-(Ethyldisulfanyl)-4-(N-methylformamido)pent-3-en-1-yl((1R,4R)-4-(N-methylbenzo[c][1,2,5]oxadiazole-5-carboxamido)cyclohexyl) glutarate (7a, Figure S7). 43.2% yield as a white solid. Mp: 78.8–79.5 °C; 1H-NMR (CDCl3, ppm): 1.27 (t, 3H, J = 7.3 Hz), 1.99, 1.97 (2s, 3H), 1.94–1.62 (m, 8H), 2.12–1.99 (m, 2H), 2.44–2.35 (m, 4H), 2.63 (q, 2H, J = 7.3 Hz), 3.04–2.87 (m, 8H), 4.70–4.59, 3.51 (m, 1H), 4.24 (t, 2H, J = 6.7 Hz), 7.41 (d, 1H, J = 8.1 Hz); 13C-NMR (CDCl3, ppm): 172.6, 172.2, 168.6, 162.3, 161.2, 148.4, 139.7, 136.5, 130.6, 117.6, 114.5, 113.6, 71.8, 62.0, 51.9, 33.3, 33.0, 31.8, 30.3, 29.6, 29.6, 29.0, 29.0, 28.2, 26.9, 19.8, 18.4, 14.0. HRMS (ESI+) m/z [M + H]+ calculated for C28H38N4O7S2: 607.2182; found: 607.2255.
S-3-(Propyldisulfanyl)-4-(N-methylformamido)pent-3-en-1-yl((1R,4R)-4-(N-methylbenzo[c][1,2,5]oxadiazole-5-carboxamido)cyclohexyl) glutarate (7b, Figure S8). 40.7% yield as a white solid. Mp: 78.0–79.3 °C; 1H-NMR (CDCl3, ppm): 0.96 (t, 3H, J = 7.3 Hz), 1.94–1.23 (m, 10H), 2.00, 1.97 (2s, 3H), 2.13–2.11 (m, 2H), 2.42–2.26 (m, 4H), 2.58 (t, 2H, J = 6.2 Hz), 3.04–2.88 (m, 8H), 4.70–4.56, 3.50 (m, 1H), 4.25 (t, 2H, J = 5.6 Hz), 7.42 (d, 1H, J = 8.9 Hz), 7.98–7.83 (m, 3H); 13C-NMR (CDCl3, ppm): 172.7, 172.3, 168.8, 162.3, 161.2, 148.4, 139.7, 136.6, 130.7, 117.5, 114.6, 113.5, 71.8, 62.1, 51.9, 41.1, 33.3, 33.1, 30.4, 29.6, 29.6, 29.0, 29.0, 28.2, 26.9, 21.9, 19.9, 18.5, 12.9. HRMS (ESI+) m/z [M + H]+ calculated for C29H40N4O7S2: 621.2338; found: 621.2411.
S-3-(Butyldisulfanyl)-4-(N-methylformamido)pent-3-en-1-yl((1R,4R)-4-(N-methylbenzo[c][1,2,5]oxadiazole-5-carboxamido)cyclohexyl) glutarate (7c, Figure S9). 38.3% yield as a white solid. Mp: 67.6–68.4 °C; 1H-NMR (CDCl3, ppm): 0.91 (t, 3H, J = 7.6 Hz), 2.00–1.25 (m, 12H), 2.03, 2.00 (2s, 3H), 2.15–2.09 (m, 2H), 2.40–2.26 (m, 4H), 2.61 (t, 2H, J = 7.3 Hz), 3.03–2.87 (m, 8H), 4.72–4.53, 3.51 (m, 1H), 4.25 (t, 2H, J = 7.0 Hz), 7.42 (d, 1H, J = 8.7 Hz), 7.94–7.83 (m, 3H); 13C-NMR (CDCl3, ppm): 172.7, 172.3, 168.6, 162.4, 161.2, 148.3, 139.8 136.6, 130.7, 117.5, 114.6, 113.7, 71.6, 62.1, 52.0, 39.0, 33.4, 33.3, 31,7, 30.4, 29.6, 29.6, 29.0, 29.0, 28.3, 26.9, 21.5, 20.0, 18.5, 13.6. HRMS (ESI+) m/z [M + H]+ calculated for C30H42N4O7S2: 635.2495; found: 635.2568.
S-3-(Amyldisulfanyl)-4-(N-methylformamido)pent-3-en-1-yl((1R,4R)-4-(N-methylbenzo[c][1,2,5]oxadiazole-5-carboxamido)cyclohexyl) glutarate (7d, Figure S10). 45.5% yield as a white solid. Mp: 69.3–70.3 °C; 1H-NMR (CDCl3, ppm): 0.89 (t, 3H, J = 6.8 Hz), 1.94–1.28 (m, 14H), 2.00, 1.97 (2s, 3H), 2.14–2.09 (m, 2H), 2.41–2.27 (m, 4H), 2.60 (t, 2H, J = 5.8 Hz), 3.04–2.88 (m, 8H), 4.71–4.59, 3.50 (m, 1H), 4.24 (t, 2H, J = 6.8 Hz), 7.42 (d, 1H, J = 8.4 Hz), 7.94–7.84 (m, 3H); 13C-NMR (CDCl3, ppm): 172.7, 172.2, 168.8, 162.4, 161.2, 148.5, 139.8, 136.6, 130.4, 117.4, 114.6, 113.7, 71.9, 62.1, 51.9, 39.3, 33.4, 33.1, 31.9, 30.5, 30.2, 29.6, 29.6, 29.0, 29.0, 28.3, 26.9, 22.2, 19.2, 18.5, 13.9. HRMS (ESI+) m/z [M + H]+ calculated for C31H44N4O7S2: 649.2651; found: 649.2724.
S-3-(Isoamyldisulfanyl)-4-(N-methylformamido)pent-3-en-1-yl((1R,4R)-4-(N-methylbenzo[c][1,2,5]oxadiazole-5-carboxamido)cyclohexyl) glutarate (7e, Figure S11). 45.0% yield as a white solid. Mp: 74.0–75.4 °C; 1H-NMR (CDCl3, ppm): 0.89 (d, 6H, J = 6.5 Hz), 1.97–1.47 (m, 11H), 2.00,1.97 (2s, 3H), 2.15–2.09 (m, 2H), 2.41–2.26 (m, 4H), 2.62 (t, 2H, J = 6.7 Hz), 3.04–2.88 (m, 8H), 4.71–4.57, 3.51 (m, 2H), 4.24 (d, 2H, J = 6.4 Hz), 7.42 (d, 1H, J = 8.1 Hz), 9.97–7.83 (m, 3H); 13C-NMR (CDCl3, ppm): 172.7, 172.3, 168.6, 162.4, 161.3, 148.5, 139.7, 136.5, 130.8, 117.4, 114.6, 113.6, 71.8, 62.1, 51.9, 37.6, 37.4, 33.4, 33.1, 31.9, 30.2, 29.6, 29.6, 29.0, 29.0, 28.3, 28.3, 26.9, 22.2, 20.0, 18.5. HRMS (ESI+) m/z [M + H]+ calculated for C31H44N4O7S2: 649.2651; found: 649.2723.
S-3-(Benzyldisulfanyl)-4-(N-methylformamido)pent-3-en-1-yl((1R,4R)-4-(N-methylbenzo[c][1,2,5]oxadiazole-5-carboxamido)cyclohexyl) glutarate (7f, Figure S12). 57.5% yield as a white solid. Mp: 72.9–73.8 °C; 1H-NMR (CDCl3, ppm): 1.91–1.25 (m, 8H), 1.94, 1.91 (2s, 3H), 2.10–2.03 (m, 2H), 2.41–2.25 (m, 4H), 3.00–2.75 (m, 8H), 4.69–4.59, 3.50 (m, 1H), 3.86 (s, 2H), 4.15 (t, 2H, J = 7.1 Hz), 7.42–7.28 (m, 5H), 7.97–7.83 (m, 3H). 13C-NMR (CDCl3, ppm): 172.6, 172.3, 168.8, 162.4, 161.3, 148.4, 139.7, 136.1, 135.8, 130.4, 129.2, 129.2, 128.5, 128.5, 127.7, 117.7, 114.6, 113.6, 71.8, 62.0, 51.9, 44.2, 33.3, 33.3, 31.9, 30.4, 29.5, 29.5, 28.7, 28.7, 26.9, 19.9, 18.4. HRMS (ESI+) m/z [M + H]+ calculated for C33H40N4O7S2: 669.2338; found: 669.2411.

4. Conclusions

In this paper, a series of brain-targeted prodrugs of LCX001 was synthesized and evaluated. The in vitro trials showed that prodrugs 7e, 7d, 7f possessed a certain stability in plasma which was propitious to get sufficient time to be delivered across the BBB. Moreover, prodrugs 7e, 7d, 7f were quickly decomposed in brain homogenate by the disulfide reductase, indicating the “lock-in” effect of these compounds after they entered the central nervous system (CNS). In vivo, prodrug 7e decreased the peripheral distribution of LCX001 and significantly increased brain distribution of LCX001 after i.v. administration. As a result, a lower dosage of prodrug 7e could give the same effect as the parent drug LCX001 on reversing opiate-induced respiratory depression, which was helpful in promoting the therapeutic efficacy and safety. More significantly, this prodrug also provides an important reference value to evaluate the clinical outlook of ampakine compounds.

Supplementary Materials

Supplementary materials can be accessed at: https://www.mdpi.com/1420-3049/21/4/488/s1.

Acknowledgments

Funding for this study was provided by the Integrated Drug Discovery Technology Platform (2012ZX09301003-001-006) of National Science and Technology Major Projects for “Major New Drugs Innovation and Development” in China.

Author Contributions

X.-B.Z., Z.Y. and S.L. conceived the project; D.X. and X.-B.Z. designed the experiments and executed the chemical synthesis; F.-H.M. and J.-Q.L. carried out the pharmacokinetics experiments; W.D. and Z.Y. performed the pharmacodynamic experiments; D.X. wrote the paper. All authors discussed the results and commented on the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AMPAAmino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
BBBBlood-brain barrier
EPSPExcitatory postsynaptic potential
FDAFood and Drug Administration
INDInvestigational new drug
ADHDAttention-deficit hyperactivity disorder
TDSThiamine disulfide system
EDCI1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride
HOBTN-Hydroxybenzotriazole
DMAP4-Dimethylaminopyridine
TEATriethylamine
DCMDichloromethane
rtRoom temperature
MRTMean residence times
RDRespiratory depression
scSubcutaneous injection
ivIntravenous injection
NMRNuclear Magnetic Resonance
LCLiquid chromatography
MSMass spectrometry
ESIElectrospray ionization
CADCollision-activated dissociation
CECollision energies
USAUnited States of America
TMSTetramethylsilane

References

  1. Swarm, R.A.; Karanikolas, M.; Kalauokalani, D. Pain treatment in the perioperative period. Curr. Probl. Surg. 2001, 38, 835–920. [Google Scholar] [CrossRef] [PubMed]
  2. Desrosiers, G. When opioid analgesia kills. Perspect. Infirm. 2006, 4, 6–9. [Google Scholar] [PubMed]
  3. Lötsch, J.; Dudziak, R.; Freynhagen, R.; Marschner, J.; Geisslinger, G. Fatal respiratory depression after multiple intravenous morphine injections. Clin. Pharmacokinet. 2006, 45, 1051–1060. [Google Scholar] [CrossRef] [PubMed]
  4. Dahan, A.; Aarts, L.; Smith, T.W. Incidence, reversal, and prevention of opioid-induced respiratory depression. Anesthesiology 2010, 112, 226–238. [Google Scholar] [CrossRef] [PubMed]
  5. Jammal, W.; Gown, G. Opioid prescribing pitfalls: Medicolegal and regulatory issues. Aust. Prescr. 2015, 38, 198–203. [Google Scholar] [CrossRef] [PubMed]
  6. Davis, C.S.; Southwell, J.K.; Niehaus, V.R.; Walley, A.Y.; Dailey, M.W. Emergency medical services naloxone access: A national systematic legal review. Acad. Emerg. Med. 2014, 21, 1173–1177. [Google Scholar] [CrossRef] [PubMed]
  7. Rekling, J.C.; Feldman, J.L. PreBotzinger complex and pacemaker neurons: Hypothesized site and kernel for respiratory rhythm generation. Annu. Rev. Physiol. 1998, 60, 385–405. [Google Scholar] [CrossRef] [PubMed]
  8. Shao, X.M.; Ge, Q.; Feldman, J.L. Modulation of AMPA receptors by cAMP-dependent protein kinase in preBötzinger complex inspiratory neurons regulates respiratory rhythm in the rat. J. Physiol. 2003, 547, 543–553. [Google Scholar] [CrossRef] [PubMed]
  9. Kc, P.; Martin, R.J. Role of central neurotransmission and chemoreception on airway control. Respir. Physiol. Neurobiol. 2010, 173, 213–222. [Google Scholar] [CrossRef] [PubMed]
  10. Pace, R.W.; Del Negro, C.A. AMPA and metabotropic glutamate receptors cooperatively generate inspiratory-like depolarization in mouserespiratory neurons in vitro. Eur. J. Neurosci. 2008, 28, 2434–2342. [Google Scholar] [CrossRef] [PubMed]
  11. Ren, J.; Poon, B.Y.; Tang, Y.; Funk, G.D.; Greer, J.J. Ampakines alleviate respiratory depression in rats. Am. J. Respir. Crit. Care Med. 2006, 174, 1384–1391. [Google Scholar] [CrossRef] [PubMed]
  12. Ren, J.; Ding, X.; Funk, G.D.; Greer, J.J. Ampakine CX717 protects against fentanyl-induced respiratory depression and lethal apnea in rats. Anesthesiology 2009, 110, 1364–1370. [Google Scholar] [CrossRef] [PubMed]
  13. Greer, J.J.; Ren, J. Ampakine therapy to counter fentanyl-induced respiratory depression. Respir. Physiol. Neurobiol. 2009, 168, 153–157. [Google Scholar] [CrossRef] [PubMed]
  14. Oertel, B.G.; Felden, L.; Tran, P.V.; Bradshaw, M.H.; Angst, M.S.; Schmidt, H.; Johnson, S.; Greer, J.J.; Geisslinger, G.; Varney, M.A.; et al. Selective antagonism of opioid-induced ventilatory depression by an ampakine molecule in humans without loss of opioid analgesia. Clin. Pharmacol. Ther. 2010, 87, 204–211. [Google Scholar] [CrossRef] [PubMed]
  15. Ren, J.; Ding, X.; Greer, J.J. Respiratory depression in rats induced by alcohol and barbiturate and rescue by ampakine CX717. J. Appl. Physiol. 2012, 113, 1004–1011. [Google Scholar] [CrossRef] [PubMed]
  16. Ren, J.; Lenal, F.; Yang, M.; Ding, X.; Greer, J.J. Coadministration of the AMPAKINE CX717 with propofol reduces respiratory depression and fatal apneas. Anesthesiology 2013, 118, 1437–1445. [Google Scholar] [CrossRef] [PubMed]
  17. ElMallah, M.K.; Pagliardini, S.; Turner, S.M.; Cerreta, A.J.; Falk, D.J.; Byrne, B.J.; Greer, J.J.; Fuller, D.D. Stimulation of Respiratory Motor Output and Ventilation in a Murine Model of Pompe Disease by Ampakines. Am. J. Respir. Cell Mol. Biol. 2015, 53, 326–335. [Google Scholar] [CrossRef] [PubMed]
  18. Ren, J.; Ding, X.; Greer, J.J. Ampakines enhance weak endogenous respiratory drive and alleviate apnea in perinatal rats. Am. J. Respir. Crit. Care Med. 2015, 191, 704–710. [Google Scholar] [CrossRef] [PubMed]
  19. Partin, K.M. AMPA receptor potentiators: From drug design to cognitive enhancement. Curr. Opin. Pharmacol. 2015, 20, 46–53. [Google Scholar] [CrossRef] [PubMed]
  20. Lippa Arnold, S. “Strategic Initiatives in Respiratory Disorders and Key Objectives”. In Proceedings of the Rodman and Renshaw 16th Annual Global Investment Conference, New York, NY, USA, 8–11 September 2014.
  21. Street, L.; Mueller, R.; Lee, S. Bicyclic Amide Derivatives for the Treatment of Respiratory Depression. International Patent 2008143963, 27 November 2008. [Google Scholar]
  22. Mueller, R.; Street, L. Bicyclic Amides for Enhancing Glutamatergic Synaptic Responses. U.S. Patent 8263591, 11 September 2012. [Google Scholar]
  23. Toyoaki, I.; Takashi, S.; Hiroshi, I.; Takashi, K.; Teruomi, I. Drug delivery to the brain. DOPA prodrugs based on a ring-closure reaction to quaternary thiazolium compounds. Int. J. Pharm. 1995, 116, 51–63. [Google Scholar]
  24. Fanm, W.; Wu, Y.; Li, X.K.; Yao, N.; Li, X.; Yu, Y.G.; Hai, L. Design, synthesis and biological evaluation of brain-specific glucosyl thiamine disulfide prodrugs of naproxen. Eur. J. Med. Chem. 2011, 46, 3651–3661. [Google Scholar]
  25. Zhao, Y.; Qu, B.; Wu, X.; Li, X.; Liu, Q.; Jin, X.; Guo, L.; Hai, L.; Wu, Y. Design, synthesis and biological evaluation of brain targeting L-ascorbic acid prodrugs of ibuprofen with “lock-in” function. Eur. J. Med. Chem. 2014, 82, 314–323. [Google Scholar] [CrossRef] [PubMed]
  26. Quan, W. Investigation on Receptor Mechanisms Underlying Powerful Antinociception and Low Addiction of Novel. Ph.D. thesis, Academy of Military Medical Sciences, Beijing, China, 2011. Available online: http://www.cnki.net/ (accessed on 8 April 2016). [Google Scholar]
  • Sample Availability: Samples of the compounds 713 are available from the authors.
Figure 1. Structure of benzamide derivatives, LCX001, thiamine disulfide, DOPA prodrug, naproxen prodrug, ibuprofen prodrug and the designed prodrugs of LCX001.
Figure 1. Structure of benzamide derivatives, LCX001, thiamine disulfide, DOPA prodrug, naproxen prodrug, ibuprofen prodrug and the designed prodrugs of LCX001.
Molecules 21 00488 g001
Figure 2. Sequential metabolism and brain “lock-in” pathways of brain-targeted thiamine disulfide prodrugs of LCX001.
Figure 2. Sequential metabolism and brain “lock-in” pathways of brain-targeted thiamine disulfide prodrugs of LCX001.
Molecules 21 00488 g002
Scheme 1. General synthetic route for synthesis of compounds 7a7f. Reagents and conditions: (i) iodomethane, 50 °C, 2 h, 96.2%; (ii) Na2S2O3·5H2O , ethanol/H2O, 110 °C, 7 h; (iii) NaOH, H2O, rt, 1 h, two steps, 13.3%–70.5%; (iv) glutaric anhydride, DMAP, DCM, 50 °C, 5 h; (v) LCX001, DMAP, HOBT, EDCI, TEA, DCM, 50 °C, 5 h, 38.3%–57.5%.
Scheme 1. General synthetic route for synthesis of compounds 7a7f. Reagents and conditions: (i) iodomethane, 50 °C, 2 h, 96.2%; (ii) Na2S2O3·5H2O , ethanol/H2O, 110 °C, 7 h; (iii) NaOH, H2O, rt, 1 h, two steps, 13.3%–70.5%; (iv) glutaric anhydride, DMAP, DCM, 50 °C, 5 h; (v) LCX001, DMAP, HOBT, EDCI, TEA, DCM, 50 °C, 5 h, 38.3%–57.5%.
Molecules 21 00488 sch001
Figure 3. Deliverence of LCX001 by the prodrugs in brain homogenate.
Figure 3. Deliverence of LCX001 by the prodrugs in brain homogenate.
Molecules 21 00488 g003
Figure 4. Concentration curve of LCX001 in plasma versus time after administration of LCX001 and prodrug 7e. Error bars show the value of SD (n = 3).
Figure 4. Concentration curve of LCX001 in plasma versus time after administration of LCX001 and prodrug 7e. Error bars show the value of SD (n = 3).
Molecules 21 00488 g004
Figure 5. Concentration curve of LCX001 in brain versus time after administration of LCX001 and prodrug 7e. Error bars show the value of SD (n = 3).
Figure 5. Concentration curve of LCX001 in brain versus time after administration of LCX001 and prodrug 7e. Error bars show the value of SD (n = 3).
Molecules 21 00488 g005
Table 1. Metabolic stability of compounds 7a7f in mice plasma extract and brain homogenate at 37 °C. Molecules 21 00488 i001
Table 1. Metabolic stability of compounds 7a7f in mice plasma extract and brain homogenate at 37 °C. Molecules 21 00488 i001
CompoundRBiological MatrixKinetic Constants
Ke (min−1) (×10−2)t1/2 (min)
7a Molecules 21 00488 i002Plasma31.12.23
Brain19.93.48
7b Molecules 21 00488 i003Plasma7.069.82
Brain6.8210.17
7c Molecules 21 00488 i004Plasma6.7710.24
Brain4.0517.10
7d Molecules 21 00488 i005Plasma4.5015.41
Brain6.3410.93
7e Molecules 21 00488 i006Plasma4.3415.97
Brain3.6918.78
7f Molecules 21 00488 i007Plasma4.4815.47
Brain5.7811.99
Table 2. Pharmacokinetic parameters of LCX001 in plasma after administration of LCX001 and prodrug 7e (n = 3).
Table 2. Pharmacokinetic parameters of LCX001 in plasma after administration of LCX001 and prodrug 7e (n = 3).
CompoundCmax (ng/mL)AUC0-t (h·ng/mL)Tmax (h)t1/2 (h)MRT0-t (h)
LCX001833.67 ± 79.85951.66 ± 115.520.080.53 ± 0.171.08 ± 0.04
7e590.36 ± 99.90 *384.16 ± 39.21 *0.080.48 ± 0.200.83 ± 0.03 *
* p < 0.05 with respect to naked LCX001.
Table 3. Pharmacokinetic parameters of LCX001 in brain after administration of LCX001 and prodrug 7e (n = 3).
Table 3. Pharmacokinetic parameters of LCX001 in brain after administration of LCX001 and prodrug 7e (n = 3).
CompoundCmax (ng/g)AUC0-t (h·ng/g)Tmax (h)t1/2 (h)MRT0-t (h)
LCX001609.40 ± 73.66504.32 ± 204.710.080.59 ± 0.080.70 ± 0.22
7e454.53 ± 74.691125.49 ± 336.57 *0.172.50 ± 0.42 *2.30 ± 0.73 *
* p < 0.05 with respect to naked LCX001.
Table 4. Mortality of opiate poisoned mice after administration of LCX001 and prodrug 7e.
Table 4. Mortality of opiate poisoned mice after administration of LCX001 and prodrug 7e.
ExperimentTreatmentTest MaterialnROUTEDose (mg/kg)Death Number (n)Death Rate
RD030418, 15 mg/kg, scSaline10iv 880%
RD030418, 15 mg/kg, scLCX00110iv10220%
RD030418, 15 mg/kg, sc7e10iv3000%
RD030418, 15 mg/kg, sc7e10iv1000%
RD030418, 15 mg/kg, sc7e20iv3420%
RD030418, 15 mg/kg, sc7e10iv1330%
RD030418, 15 mg/kg, sc7e10iv0.5550%
RD—Respiratory depression; sc—subcutaneous injection, iv—intravenous injection.
Table 5. Mass spectrometric conditions for the compounds.
Table 5. Mass spectrometric conditions for the compounds.
[M + H]+Precursor ionProduct ionCE (V)
LCX001276.2178.220
Glipizide446.0321.020
7a607.5258.225
7b621.5258.226
7c635.5258.227
7d649.5258.229
7e649.5258.229
7f669.5258.229

Share and Cite

MDPI and ACS Style

Xiao, D.; Meng, F.-H.; Dai, W.; Yong, Z.; Liu, J.-Q.; Zhou, X.-B.; Li, S. Design, Synthesis and Biological Evaluation of Brain-Targeted Thiamine Disulfide Prodrugs of Ampakine Compound LCX001. Molecules 2016, 21, 488. https://doi.org/10.3390/molecules21040488

AMA Style

Xiao D, Meng F-H, Dai W, Yong Z, Liu J-Q, Zhou X-B, Li S. Design, Synthesis and Biological Evaluation of Brain-Targeted Thiamine Disulfide Prodrugs of Ampakine Compound LCX001. Molecules. 2016; 21(4):488. https://doi.org/10.3390/molecules21040488

Chicago/Turabian Style

Xiao, Dian, Fan-Hua Meng, Wei Dai, Zheng Yong, Jin-Qiu Liu, Xin-Bo Zhou, and Song Li. 2016. "Design, Synthesis and Biological Evaluation of Brain-Targeted Thiamine Disulfide Prodrugs of Ampakine Compound LCX001" Molecules 21, no. 4: 488. https://doi.org/10.3390/molecules21040488

Article Metrics

Back to TopTop