Next Article in Journal
Preparation and Characterization of Natural Silk Fibroin Hydrogel for Protein Drug Delivery
Previous Article in Journal
An In Situ Formation of Ionic Liquid for Enrichment of Triazole Fungicides in Food Applications Followed by HPLC Determination
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identification of New Non-BBB Permeable Tryptophan Hydroxylase Inhibitors for Treating Obesity and Fatty Liver Disease

1
Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
2
JD Bioscience, 208 beon-gil, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Korea
3
Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
4
Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
5
Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, India
6
Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
7
Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
8
Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34141, Korea
9
Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(11), 3417; https://doi.org/10.3390/molecules27113417
Submission received: 31 March 2022 / Revised: 23 May 2022 / Accepted: 23 May 2022 / Published: 25 May 2022

Abstract

:
Serotonin (5-hydroxytryptophan) is a hormone that regulates emotions in the central nervous system. However, serotonin in the peripheral system is associated with obesity and fatty liver disease. Because serotonin cannot cross the blood-brain barrier (BBB), we focused on identifying new tryptophan hydroxylase type I (TPH1) inhibitors that act only in peripheral tissues for treating obesity and fatty liver disease without affecting the central nervous system. Structural optimization inspired by para-chlorophenylalanine (pCPA) resulted in the identification of a series of oxyphenylalanine and heterocyclic phenylalanine derivatives as TPH1 inhibitors. Among these compounds, compound 18i with an IC50 value of 37 nM was the most active in vitro. Additionally, compound 18i showed good liver microsomal stability and did not significantly inhibit CYP and Herg. Furthermore, this TPH1 inhibitor was able to actively interact with the peripheral system without penetrating the BBB. Compound 18i and its prodrug reduced body weight gain in mammals and decreased in vivo fat accumulation.

1. Introduction

Serotonin (5-hydroxytryptamine [5-HT]) is an ancient biochemical that acts in the central nervous system and peripheral nervous system [1]. In the central nervous system, 5-HT is a neurotransmitter, which affects mood, appetite, sleep, and memory [2,3,4,5]. An imbalance in the serotonin system in the central nervous system has been implicated in a multitude of neuropsychiatric diseases. Recently, the new function regarding peripheral serotonin in energy homeostasis ranging from the endocrine regulation by gut-derived serotonin to the autocrine/paracrine regulation by adipocyte-derived serotonin was reported [6]. Serotonin plays an important role in metabolic regulation in peripheral tissues and is emerging as a possible target for anti-obesity and fatty liver disease treatment. Serotonin is synthesized from tryptophan by the sequential action of two enzymes, tryptophan hydroxylase (TPH) and aromatic amino acid decarboxylase with TPH catalyzing the rate-limiting step (Figure 1).
Oh et al. [7] reported that serotonin regulates white and brown adipose tissue function. Mice with inducible TPH1 KO in adipose tissues exhibit inhibition of lipogenesis in epididymal white adipose tissue (WAT), induction of browning in inguinal WAT, and activation of adaptive thermogenesis in brown adipose tissue (BAT).
The inability of serotonin to cross the Blood Brain Barriers (BBB) enforces the dualistic character of the serotonin system by creating two physiologically separated serotonin pools in the body. These characteristics of serotonin and KO study results prompted us to develop a TPH1 inhibitor that only acts in peripheral tissue for treating obesity and fatty liver disease.
Several tryptophan hydroxylase type I (TPH1) inhibitors have been reported in the literature and patents. For example, para-chlorophenylalanine (pCPA), [8] also known as fenclonine, is a classic TPH1 inhibitor that has been developed to treat carcinoid syndrome. However, the in vitro binding activity of pCPA to TPH1 is very weak (IC50 > 50 µM). Additionally, pCPA causes side effects such as depression because it passes through the central nervous system. Recently, telotristat ethyl was marketed as a TPH1 inhibitor for carcinoid syndrome [9]. Scientists at Karos Pharmaceuticals have reported that spirocyclic TPH1 inhibitors such as telotristat ethyl can be used to treat Pulmonary Arterial Hypertension (PAH) and Inflammatory Bowel Disease (IBD) [10].
Herein, we attempt to identify new TPH1 inhibitors starting from pCPA by introducing a non-BBB permeable moiety. The design, synthesis, and biological evaluation of a diverse suite of phenylalanine derivatives for preventing obesity and fatty liver disease are presented.

2. Results and Discussion

2.1. Chemistry

Tyrosine, which is similar to pCPA, was selected as the starting material because it is suitable for derivatization. The general methods for the synthesis of oxy-phenylalanine derivatives are outlined in Scheme 1. Commercially available L-tyrosine (1) was first esterified, and Boc-anhydride was then introduced to provide Boc protection, which yielded compound 2. Compound 2 was subsequently coupled with 7-bromo-4-chlorothieno[3,2-d] pyrimidine (3) to yield compound 4. Compound 4 was treated with NaOH and subjected to Boc deprotection with HCl (4 M) in 1,4-dioxane to give compound 6. Compound 4 also underwent Suzuki coupling with 3-hydroxyphenylboronic acid (5a) and 4-hydroxyphenylboronic acid (5b) in the presence of a palladium catalyst to afford compounds 7a and 7b; these compounds were hydrolyzed and underwent Boc deprotection to give compounds 9a and 9b. Compounds 7a and 7b also underwent the Mitsunobu reaction with benzyl alcohol to afford compounds 10a and 10b, which were treated with NaOH and underwent Boc-deprotection with HCl (4 M) in 1,4-dioxane to give compounds 11a and 11b. Compounds 15a and 15b were also synthesized, as shown in Scheme 1. Thienopyrimidine (3) was ammonified to afford compound 13a, and thienopyrimidine (3) was treated with aqueous NaOH to afford compound 13b. Compounds 12 and 13b were coupled via Suzuki coupling to afford compounds 14a and 14b. Triflic anhydride and bis(pinacolato) diboron were used to prepare compound 12 from compound 2. Compounds 14a and 14b were then hydrolyzed, and subsequent acid deprotection afforded compounds 15a and 15b.
Amino and oxy derivatives were synthesized by substitution at position-4 of the thienopyrimidine ring; the general synthesis is shown in Scheme 2. Commercially available 7-bromo-4-chlorothieno[3,2-d]pyrimidine 3 was condensed with [1,1’-biphenyl]-4-ylmethanamine (16a) and (R)-1-(naphthalen-2-yl)ethan-1-amine (16b), phenyl 2,2,2-trifluoroethan-1-ol (16c), and substituted phenyl 2,2,2-trifluoroethan-1-ol (16d–i). The condensates subsequently underwent Suzuki coupling with ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propanoate (12) to yield compounds 17a–i, which were subsequently saponified and underwent acidic deprotection to afford several amine hydrochlorides (compounds 18a–i). Compound 17i underwent tert-butyl deprotection to give compound 19, which was treated with ammonium hydroxide to give an amine, compound 20. Hippuric acid was treated with compound 20 to produce a hippurate salt, compound 21.
The substituted benzaldehydes (22ch) were treated with (trifluoromethyl)trime thylsilane and tetrabutylammonium fluoride which yielded compounds 16c and 23dh. Compounds 23dh underwent Suzuki coupling with phenyl boronic acid (24a), furan-3-ylboronic acid (24b), and 5,6-dihydro-2H-pyran-3-yl)boronic acid (24c) which yielded compounds 16dh. Commercially available (R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethan-1-ol (16i) was used to synthesize compound 18i.
The synthesis of compound 30 is shown in Scheme 3. Compound 25 was cyclized with urea and then brominated to afford compound 26 which was subsequently chlorinated with POCl3 to obtain compound 27. Compound 27 was further treated with compound 16i and p-methoxybenzylamine to afford compound 28. TFA deprotection afforded compound 29, which was then hydrolyzed; acidic deprotection of 29 afforded compound 30.

2.2. Biological Evaluations

The in-house screening revealed that thienopyrimidine tyrosine (derivative 6) was active in the inhibition of TPH1, with an inhibition of 64% at 100 µM. Consequently, the core structure of thienopyrimidine tyrosine (derivative 6) was selected as the starting point for the synthesis, evaluating the structure-activity relationships (SAR), and optimization of TPH1 inhibitors. The ability of the synthesized compounds to inhibit TPH1 at 100 µM and 1 µM, respectively, was evaluated. In the evaluation, pCPA and LP533401 (4-[2-amino-6-[2,2,2-trifluoro-1-(3’-fluoro[1,1’-biphenyl]-4-yl)ethoxy]-4-pyrimidinyl]-L-phenylalanine) were used as references.
The optimized parameters for the oxy-heteroaryl phenylalanine derivatives of compound 6 are summarized in Table 1. Compound 6 exhibited an inhibition that was greater than 60%. When substituents were introduced at the para position (compounds 9a and 11a), little loss of the inhibitory activity occurred (45% and 58% inhibition at 100 µM, respectively). Compounds 9b and 11b (82% and 94% inhibition at 100 µM, respectively) showed better inhibitory activity than compounds 9a and 11a, respectively.
Based on these results, computational molecular docking studies were conducted to improve the molecular level understanding of effective binding to the active site; these studies are fundamental for facilitating further structural modification. For the docking studies, the high-resolution structure of human TPH1 (Protein Data Bank access code 3HF8; chain A) was used in AutoDock4.2 [11,12]. The standard protocols and parameters described in our previous work were used to pre-process the structures [13]. The molecular docking results showed that compound 11b interacted with the catalytic residues of TPH1 (Arg257 and Thr265). In addition, the hydrophobic tail of compound 11b was bound to the exterior of the active site of TPH1 with weak interactions and unable to block the entrance of the TPH1 active site. Such binding of the hydrophobic tail of compound 11b is leaving the possibility of substrate binding with the TPH1 in a competitive manner (Figure 2). We hypothesized that the interaction of the hydrophobic tail of synthesized compounds at the entrance of the active site of TPH1 might prevent the substrate access the active site of TPH1 which might lead to better inhibition. Based on these observations and our hypothesis, we first replaced oxy-phenylalanine to produce compounds 15a, 15b, and 18a. Although compounds 15a and 15b showed weak activity, biphenyl methyl amine (18a) was a promising TPH1 inhibitor with an inhibition of 90% at 100 µM.
Based on these results, we explored a wide range of substituents on position-4 of the thienopyrimidine core to enhance the inhibitory activity; the results are summarized in Table 2. Naphthalene ethane amine (18b) showed less inhibition than compound 18a. Attempts were made to introduce benzyloxy derivatives at position-4 of thienopyrimidine. However, these derivatives were unstable under the acidic conditions used for Boc deprotection, as evidenced by the cleavage of the benzyl moiety to form compound 15b. To improve the stability of these derivatives, attempts were made to synthesize 18c, 18d, and 18e, which have a CF3-substituted benzyloxy moiety. The molecular docking study shows that the CF3 group interacted with Leu236 and Phe241 of TPH1. Contrastingly, compound 18c without a substituent and compound 18d with a phenyl substituent at the para position on the phenyl ring completely lost their activity. However, when the substituent was in the ortho position (compound 18e), the percentage inhibition dramatically improved at a concentration of 1 µM. Although the interactions among compound 18e and the catalytic and key residues of TPH1 were enhanced, the hydrophobic tail of compound 18e partially occupied the entrance of the active site of TPH1. This observation suggests that further modifications are required. Halogens are widely used in drug design to improve the drug-target binding affinity and slow drug metabolism; therefore, we introduced chlorine at the para position of the benzyloxy moiety. However, the introduction of chlorine in 18e yielded compound 18f, which showed a loss in the percentage inhibition. Based on the molecular docking simulation, the R isomer of the trifluoroethoxy linker was also introduced as a substituent. This approach resulted in the design and synthesis of compounds 18g, 18h, and 18i with furyl, dihydropyran, and methyl pyrazole substituents. Compared with compound 18g, compounds 18h and 18i showed a higher percentage inhibition, with IC50 values of 263 nM and 37 nM, respectively.
To optimize the binding of the CF3 group and thienopyrimidine ring with TPH1, compound 18i was synthesized as an R-enantiomer with a modified hydrophobic tail, which was capable of effectively inhibiting TPH1. The optimal interactions were observed among the thienopyrimidine ring and phenylalanine moiety of compound 18i and the aforementioned key and catalytic residues of TPH1.
Therefore, compound 18i is a more effective inhibitor than compounds 18e and 18h because of its increased potency, as evidenced by its IC50 value of 37 nM. It was further assumed that adding an amine group to the thienopyrimidine ring would likely confer a different potency profile to compound 18i. However, compound 30 disproved this assumption because it showed reduced potency, as evidenced by the IC50 value of 208 nM (Table 3). During this evaluation, LP533401 was used as a reference [14]. The prodrugs of compound 18i were synthesized to improve its oral absorption (Table 3). The ester of compound 18i, compound 19, exhibited nanomolar inhibitory activity, with an IC50 value of 994 nM. In addition, the hippurate salt (21) was synthesized as an oral drug for in vivo study.
Based on the in vitro data, compound 18i was selected for inhibiting the in vitro BBB permeability. Several in vitro methods and computational models have been used in drug discovery to predict the potential of a drug to penetrate the BBB [15,16,17,18,19,20,21]. The parallel artificial membrane permeation assay (PAMPA) was used to evaluate the BBB permeability of the selected compounds. The PAMPA-BBB assay is advantageous because it predicts passive penetration at the blood-brain barrier with high success, high throughput, low cost, and high reproducibility [21].
Table 4 shows that compound 18i has non-BBB permeable characteristics, with Pe ≈ 0.00 × 10−6 cm/s (i.e., Pe < 2.0), whereas pCPA exhibited a higher Pe of 1.85 × 10−6 cm/s. Therefore, compound 18i was chosen as a prototype for further investigation. Compound 18i showed good liver microsomal stability (99% of the parental microsomes remained after incubation for 30 min). Table 5 shows that compound 18i did not significantly inhibit any CYP isoforms (1A2, 2C19, 2D6, and 3A4).
Compound 18i was evaluated for its BBB ratio by iv injection and PK profile in rats as shown in Table 6. The levels of compound 18i in the plasma (2266 and 1439 ng/mL) indicate that it did not penetrate the BBB. Furthermore, the level of compound 18i in the plasma was below the quantification limit (7 ng/mL). The PK profiles of 18i were also evaluated which revealed that compound 18i had a reasonable AUC of 1.2 μg h/mL; however, 18i itself could not be absorbed into the body due to its low permeability. Therefore, 19 and 21 were introduced as the ethyl ester and hippurate salt prodrug, respectively, affording improved oral exposure, with 16% oral bioavailability.
Recent reports have shown that TPH inhibitors, such as pCPA and LP533401, protect against diet-induced obesity in vivo by reducing body weight gain and lipogenesis in adipose tissue in high-fat diet (HFD) fed mice [7,22]. Based on the pharmacological profiles of newly developed TPH1 inhibitors, we selected compound 18i and its prodrug compound 19 and 21 for further efficacy tests in HFD-induced obesity mouse models. First, we tested the effects of compound 18i in vitro by treating 3T3-L1 adipocyte differentiation and found decreased expression of key lipogenic genes, Fasn and Srebp1c, along with the adipogenic transcription factors, Pparg and Cebpa (Figure 3A), indicating an anti-adipogenic effect for compound 18i during adipocyte differentiation. To assess in vivo efficacy, we performed daily intraperitoneal injections of compound 18i to HFD-fed C57BL6 mice. We found that compound 18i treatment, as compared to vehicle, resulted in blunted weight gain accompanied by a significant reduction in epididymal white adipose tissue (eWAT) and brown adipose tissue (BAT) adipocyte size as assessed by histological examination (Figure 3B). Similar to the effects of compound 18i, compound 19 treatment attenuated body weight gain in HFD-fed mice compared to vehicle (Figure 4A). In addition to decreased adiposity, HFD-fed mice treated with compound 18i had lower fasting blood glucose levels compared to vehicle-treated mice (Figure 4B). Histological analysis of adipose tissues from compound 19-treated mice showed decreased adipocyte size in both eWAT and inguinal white adipose tissue (iWAT) (Figure 4C). iWAT from compound 19 treated mice had a higher content of mitochondrial uncoupling protein 1 (UCP1), a marker for thermogenic beige adipose tissue, as assessed by immunohistochemistry (Figure 4D). Notably, oral administration of compound 21 to HFD-fed mice dramatically decreased lipid accumulation in liver tissue compared to vehicle-treated mice (Figure 4E), as assessed by histological examination and oil red O staining, without significant changes in body and liver weights (Figure 4F,G). This effect by compound 21 treatment is consistent with our finding that gut tissue-specific deletion of TPH1 in mice leads to a reduction in liver steatosis in HFD-fed mice without affecting body and liver weights (unpublished data). Taken together, these results demonstrate the in vivo efficacy of our newly developed TPH1 inhibitors for ameliorating obesity and fatty liver diseases.
Docking studies revealed that compound 18i had optimal interactions with the catalytic and other important residues of TPH1, with predicted binding free energy of −14.33 kcal/mol, which effectively inhibited TPH1. The amino group in the phenylalanine moiety of compound 18i interacted with the catalytic residues, Arg257 and Thr265, and the TPH1 residues, Gly333 and Ser335 (Figure 5). The phenyl ring of the phenylalanine moiety interacted with His272 and Phe318 of TPH1. The phenyl ring of the phenylalanine moiety and the thienopyrimidine ring were optimally oriented because of Pro268 of TPH1, which facilitated the interaction of the thienopyrimidine ring with the key residues, Phe241 and Glu317. Additional interactions were observed between the sulfur of the thienopyrimidine ring and the Phe313 residue of TPH1. Modification of the hydrophobic tail of compound 18i improved its interactions, thereby blocking the entrance of the active site of TPH1. To summarize, compound 18i adopts a compact conformation that is expected to inhibit the binding of substrate cofactor and interferes with the binding of iron with TPH1, thereby leading to the effective inhibition of TPH1.

3. Experimental Section

3.1. Chemistry

All reported yields are isolated yields after column chromatography or crystallization. All solvents and chemicals were used as purchased without further purification. 1H NMR spectra and 13C spectra were recorded on a JEOL JNM-ECS400 spectrometer at 400 MHz for 1H NMR and 100 MHz for 13C NMR, respectively. The chemical shift (δ) is expressed in ppm relative to tetramethylsilane (TMS) as an internal standard, and CDCl3, DMSO-d6, and CD3ODwere used as solvents. The multiplicity of peaks is expressed as s (singlet), d (doublet), t (triplet), q (quartet), dd (doublet of doublets), td (triplet of doublets), qd (quartet of doublets), dt (doublet of triplets), and m (multiplet). Melting points were determined on a Melting Point M-560, purchased from Buchi. Fast atom bombardment-high-resolution mass spectrometry (FAB-HRMS) data were obtained by a JMS 700 (JEOL, Japan). The purity of all tested compounds was ≥ 95%, as estimated by high performance liquid chromatography (HPLC) analysis. Samples were analyzed on a Waters Agilent HPLC system equipped with a PDA detector and a Waters SB-C18 column (1.8 μm, 2.1 × 50 mm2). The mobile phase was used with buffer A (ultrapure H2O containing 0.1% TFA) and buffer B (chromatographic-grade CH3CN). The flow rate was 0.5 mL/min.

3.2. Synthesis of (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoic acid hydrochloride (18i)

3.2.1. Ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate (17i)

(R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethan-1-ol 16i (900 mg, 3.096 mmol) was dissolved in 4 mL of N,N-dimethylformamide and cooled to 0 °C. Sodium hydride 60% in oil (185.7 mg, 4.644 mmol) was added to the mixture and stirred for 60 min. 7-bromo-4-chloro-thieno[3,2-d]pyrimidine 3 (811.18 mg, 3.25 mmol) was added to the mixture and stirred at room temperature for 12 hr. The resulting mixture was quenched in aqueous ammonium chloride and extracted twice with ethyl acetate. The organic layer was washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under a reduced pressure to obtain a residue. The resulting residue was purified by column chromatography to obtain of the title compound (R)-7-bromo-4-(1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidine (1.5 g, 96%).
(R)-7-bromo-4-(1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy) thieno [3,2-d]pyrimidine (1.5 g, 2.978 mmol) was added to 1,4-dioxane (25 mL), and ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl) propanoate 12 (1.37 g, 3.26 mmol)), tetrakis(triphenylphosphine)palladium(0) (172.06 mg, 0.149 mmol), potassium carbonate (823.11 mg, 5.96 mmol), and water (8 mL) were sequentially added thereto while stirring. The reaction mixture was heated to 90 °C, and stirred at 90 °C for 3 h. After completion of the reaction using brine, the reaction mixture was extracted twice with ethyl acetate. The collected organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain a foamy residue which was purified by column chromatography to obtain the title compound ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate 17i (1.5 g, 70%).1H NMR (400 MHz, DMSO-d6): δ 8.70 (s, 1H), 8.67 (s, 1H), 8.24 (d, J = 2.44 Hz, 1H), 7.99 (d, J = 8.24 Hz, 2H), 7.88–7.78 (m, 2H), 7.70 (d, J = 2.14 Hz, 1H), 7.56 (dd, J = 8.54, 2.14 Hz, 1H), 7.34 (d, J = 8.24 Hz, 2H), 6.40 (d, J = 2.44 Hz, 1H), 4.32–4.25 (m, 1H), 4.11 (q, J = 5.19 Hz, 2H), 3.22–3.06 (m, 2H), 2.27 (s, 3H), 1.33 (s, 9H), 1.09 (t, J = 7.02 Hz, 3H).

3.2.2. (S)-2-Amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoro ethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoic acid hydrochloride (18i)

NaOH (50.27 mg, 1.26 mmol) was added to a solution of ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate 17i (180 mg, 0.251 mmol) in THF/water (50 mL, 3:1). The reaction mixture was stirred at ambient temperature for 24 h. The THF was removed in vacuo and the resulting solution was acidified with 1N hydrochloric acid to pH 4. More water was added (50 mL) and the aqueous solution was extracted with EtOAC (3 × 50 mL). The combined organic layer was washed with brine, dried over sodium sulfate, and concentrated. A crude product was purified by column chromatography to afford the title compound (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy) thieno [3,2-d]pyrimidin-7-yl)phenyl)propanoic acid.
Hydrogen chloride 4.0 M solution in 1,4 dioxane (5 mL) was added to a mixture of (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl) propanoic acid in ethyl acetate (10 mL) and the mixture was stirred for 12 h. The mixture was concentrated to minimum volume and the residue was collected by filtration to give (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoro ethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoic acid hydrochloride 18i (121 mg, 77%) (over two steps) as an off white solid. 1H NMR (400 MHz, DMSO-d6): δ 13.83 (s,1H), 8.70 (d, J = 1.22 Hz, 1H), 8.66 (d, J = 1.53 Hz, 1H), 8.37 (bs, 3H), 8.24 (s, 1H), 7.98 (d, J = 7.32 Hz, 2H), 7.88–7.77 (m, 1H), 7.72–7.68 (m, 1H), 7.56 (dt, J = 8.54,1.22 Hz, 1H), 7.37 (d, J = 7.63 Hz, 2H), 6.43–6.39 (m, 1H), 4.24–4.15 (m, 1H), 3.14 (d, J = 6.10 Hz, 2H), 2.27 (s, 3H); 13C NMR (100 MHz, DMSO-d6): δ 170.34, 157.49, 154.71, 149.94, 146.62, 139.73, 136.26, 134.47, 134.03, 132.74, 132.59, 130.95, 130.74, 129.65, 129.47, 128.17, 127.95, 124.97, 124.40, 107.27, 53.10, 35.46, 13.38; HRMS (FAB) m/z calcd for C27H21ClF3N5O3S [M + H]+ 587.1006, found 588.1089; LC-MS (m/z): 588.2 [M + H]; HPLC purity 99.12%; m.p. 215–217 °C.

3.3. Synthesis of ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate hydrochloride (19)

Hydrogen chloride 4.0 M solution in 1,4 dioxane (5 mL) was added to a mixture of ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate (500 mg, 0.698 mmol) in ethyl acetate (25 mL) and the mixture was stirred for 12 h. The mixture was concentrated and filtered to give title compound ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate hydrochloride 19 (405 mg, 88%) as an off white solid. 1H NMR (400 MHz, DMSO-d6): δ 8.75 (d, J = 3.05 Hz, 1H), 8.62 (d, J = 3.05 Hz, 1H), 8.26 (t, J = 2.44 Hz, 1H), 7.96 (dd, J = 7.93, 2.44 Hz, 2H), 7.89–7.82 (m, 2H), 7.72 (t, J = 2.44 Hz, 1H), 7.59 (dt, J = 8.85, 2.44 Hz, 1H), 7.32 (dd, J = 8.24, 2.14 Hz, 2H), 6.44 (t, J = 2.44 Hz, 1H), 4.02 (q, J = 7.02 Hz, 2H), 3.63–3.58 (m, 1H), 2.95–2.77 (m, 2H), 2.30 (s, 3H), 1.13 (t, J = 7.02 Hz, 3H); 13C NMR (100 MHz, DMSO-d6): δ 168.95, 161.37, 159.95, 135.74, 150.48, 140.66, 135.51, 135.18, 134.65, 133.27, 132.71, 131.92, 130.16, 129.75, 128.13, 128.08, 124.63, 123.83, 117.82, 107.82, 61.71, 53.08, 35.66, 13.83, 13.15; HRMS (FAB) m/z calcd for C29H25ClF3N5O3S [M + H]+ 615.1319, found 616.1399; LC-MS (m/z): 616.05 [M + H]; HPLC purity 98.8%; m.p. 102–104 °C.

3.4. Synthesis of ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate (20)

Hydrogen chloride 4.0 M solution in 1,4 dioxane ( 10 mL) was added to a mixture of ethyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate (1.2 g, 1.676 mmol) in ethyl acetate (20 mL) and the mixture was stirred for 12 h. The mixture was concentrated and the residue was dissolved in water. The pH was adjusted to 8 by aqueous ammonia and extracted with ethyl acetate. The combined organic layer was dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel column chromatography to give title compound ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate 20 (910 mg, 88%) as an off white solid. 1H NMR (400 MHz, DMSO-d6): δ 8.72 (d, J = 3.05 Hz, 1H), 8.64 (d, J = 3.05 Hz, 1H), 8.26 (t, J = 2.44 Hz, 1H), 7.94 (dd, J = 7.93, 2.44 Hz, 2H), 7.89–7.80 (m, 2H), 7.72 (t, J = 2.44 Hz, 1H), 7.59 (dt, J = 8.85, 2.44 Hz, 1H), 7.30 (dd, J = 8.24, 2.14 Hz, 2H), 6.44 (t, J = 2.44 Hz, 1H), 4.04 (q, J = 7.02 Hz, 2H), 3.62–3.55 (m, 1H), 2.96–2.78 (m, 2H), 2.30 (s, 3H), 1.12 (t, J = 7.02 Hz, 3H); 13C NMR (100 MHz, DMSO-d6): δ 173.91, 161.34, 159.98, 153.65, 150.46, 140.67, 135.48, 132.68, 130.13, 129.60, 129.50, 127.95, 127.81, 124.63, 123.85, 107.79, 60.25, 55.23, 36.70, 14.01, 13.48; HRMS (FAB) m/z calcd for C29H25ClF3N5O3S [M + H]+ 615.1319, found 616.14; LC-MS (m/z): 616.0 [M + H]; HPLC purity 99.24%; m.p. 180–182 °C.

3.5. Synthesis of ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate hippurate (21)

To a solution of ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate (330 mg, 0.536 mmol) in ethanol (50 mL) was added hippuric acid (95.98 mg, 0.536 mmol). The reaction mixture was stirred at reflux for 12 h and cooled to room temperature. The reaction mixture was concentrated under reduced pressure to obtain a bubble residue. The resulting residue was dissolved in methyl tert-butyl ether (25 mL) and added n-Hexane to form a solid which filtered to give ethyl (S)-2-amino-3-(4-(4-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)thieno[3,2-d]pyrimidin-7-yl)phenyl)propanoate hippurate 21 as a white solid. 1H NMR (400 MHz, DMSO-d6): δ 8.77 (t, J = 5.49 Hz, 1H), 8.72 (s, 1H), 8.65 (s, 1H), 8.27 (d, J = 2.44 Hz, 1H), 7.95 (d, J = 7.93 Hz, 2H), 7.89–7.80 (m, 5H), 7.72 (d, J = 2.14 Hz, 1H), 7.59 (dd, J = 8.54, 2.14 Hz, 1H), 7.57–7.44 (m, 4H), 7.31 (d, J = 8.24 Hz, 2H), 6.44 (d, J = 2.14 Hz, 1H), 4.05 (q, J = 7.02 Hz, 2H), 3.90 (d, J = 5.80 Hz, 2H), 3.66–3.58 (m, 1H), 2.96–2.80 (m, 2H), 2.31 (s, 3H), 1.09 (t, 7.02 Hz, 3H); 13C NMR (100 MHz, DMSO-d6): δ 171.44, 166.34, 161.36, 160.00, 153.67, 150.48, 140.69. 135.57, 135.51, 133.97, 132.81, 132.71, 131.34, 131.22, 130.16, 129.64, 129.52, 128.34, 128.00, 127.83, 127.22, 124.66, 123.87, 117.78, 107.82, 60.26, 55.30, 41.50, 36.66, 14.03, 13.50; HRMS (FAB) m/z calcd for C29H25ClF3N5O3S [M + H]+ 615.1319, found 615.7; LC-MS (m/z): 616.2 [M + H]+; HPLC purity 98.79%; m.p. 87–89 °C.

3.6. Biology

3.6.1. Microsomal Activity

For the liver microsomal activity, test compounds were incubated with rat and mouse’s liver microsomes, respectively, at a final concentration of 1 µM, in the presence of an NADPH regeneration system in 0.1 M potassium phosphate buffer (pH 7.4) for 30 min h at 37 °C in shaking incubator. The microsomal reaction was terminated by adding four volumes of ice-cold acetonitrile and then centrifuged at 4000 rpm for 20 min. The supernatant was collected and analyzed by a LC-MS/MS system. The percentage of the parent compound remaining was calculated by comparing peak areas.

3.6.2. CYP Inhibition Assay

Five major screening systems: P450-Glo CYP1A2 Screening System (Catalog# V9770), P450-Glo CYP2C9 Screening System (Catalog# V9790), P450-Glo CYP2C19 (Catalog# V9880), P450-Glo CYP 2D6 (Catalog# V9890), and P450-Glo CYP3A4 (Catalog# V9920) were attained from Promega Corp. (Madison, WI). α-naphthoflavone, sulfaphenazole, amitriptyline, quinidine, and ketoconazole were identified as positive control inhibitors for CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4, respectively. The CYP enzyme and substrate were combined in a potassium phosphate (KPO4) buffer and the reaction is initiated by adding an NADPH regenerating system. The volume of the mixture (12.5 μL in a 96-well plate) was combined with an equal volume of test compound solution and positive control inhibitor (12.5 μL added to make the volume 25 μL). The reaction of the NADPH regeneration system was initiated by adding a 2× concentration of NADPH solution. Then an equal volume of luciferin detection reagent was added. The plates were incubated at room temperature for 20 min and the luminescence signal was measured by a microplate reader device.

3.6.3. The hERG Inhibition Activity

The inhibition of binding affinity by small molecules was measured using the Predictor™ hERG FP kit (Thermo Fisher Scientific, Inc., Rockford, IL, USA). The procedure was carried out in accordance with the manufacturer’s instructions. Briefly, tracer (Predictor™ hERG tracer red) was prepared with dilution in the binding buffer. The binding assay was conducted in 384 well black flat-bottom microplates (Corning Life Sciences, Lowell, MA, USA) after the serial addition of 5 μL of a 4 nM tracer, 5 μL of test compounds, and a 10 μL membrane fraction containing hERG channel protein. The mixtures were incubated at room temperature for 2 h. The fluorescence polarization (FP) was measured with an excitation filter of 530 nm and an emission filter of 585 nm using a microplate reader (Infinite M1000PRO; Tecan, Mannedorf, Switzerland).

3.6.4. Blood Brain Barriers PK

The biodistribution study in plasma and brain tissue was performed using eight-week-old male SD rats (Orient Bio Inc., Seongnam, Korea). Dosing vehicles were composed of DMSO, PEG400, and deionized water (5:40:55, v/v/v). 18i compound was administrated at a dose of 5 mg/kg via intravenous (I.V.). Plasma from the inferior vena cava and whole brain tissue were collected at 0.5 and 3 h after drug administration. Brain tissues were rinsed with saline and four volumes of acetronitrile containing disopyramide were added as an internal standard for LC-MS/MS analysis. The brain samples were homogenized using a sonicator (IKA Labortechnik T10 basic ULTRA-TURRAX, Staufen, Germany) for 10 s on the ice. The plasma samples were added to nine volumes of acetonitrile and vortexed for 5 min. The samples were centrifuged (15,000 RPM, 4 °C, 10 min). The supernatant was collected and analyzed by an LC-MS/MS system.

3.6.5. Parallel Artificial Membrane Permeability Assay (PAMPA)

PAMPA was conducted with a STIRWELLTM PAMPA sandwich (pION, Inc., Billerica, MA, USA) consisting of a 96-well filter plate (0.45 μm PVDF membrane) and a 96-well polystyrene donor plate. Test compounds were prepared in 10 mM stock solution with DMSO and diluted to a working solution of 50 μM concentration using a phosphate buffered saline (PBS) buffer. To form lipid membranes in the acceptor plate, 5 μL GIT-0 lipid (pION, Inc., Billerica, MA, USA) was dropped and semi-dried on the bottom side of the 0.45 μm PVDF membrane. The donor plate was prepared by the addition of 200 μL of working solution. The acceptor plate coated with lipid membrane was then overlayed on the donor plate without an air bubble. Continuously, 200 μL of acceptor sink buffer (pION, Inc., Billerica, MA, USA) was dispensed at the inner side of the acceptor plate. The combined cassette plate was incubated for 4 h at room temperature in Gut-BoxTM (pION, Inc., Billerica, MA, USA). To acquire the peak area of range from 230 to 500 nm, absorbance scans of PBS, acceptor, donor, and working solution were carried out with an Epoch ELISA reader (BioTek Instruments Inc., Winooski, VT, USA). The PAMPA Explorer Command Software (pION, Inc., Billerica, MA, USA) was used for the analysis of the permeability of compounds.

3.6.6. Pharmacokinetic Study

The pharmacokinetic study of 18i was performed using eight-week-old male SD rats (Orient Bio Inc., Seongnam, Korea). 18i and its prodrug 21 (5 mg/kg, 2 mL/kg) were administered via intravenous and oral routes. Blood samples were collected at 0.033, 0.16, 0.5, 1, 2, 4, 6, 8, and 24 h for intravenous injection at 0.15, 0.5, 1, 2, 4, 6, 8, and 24 h for oral dosing after drug administration, and then immediately centrifuged at 10,000× g for 3 min. The plasma concentrations of 18i were determined by LC-MS/MS. The plasma concentration-time profiles and pharmacokientic parameters were analyzed by a non-compartmental method using the nonlinear least-squares regression program WinNonlin 5.3 (Pharsight, Mountain View, CA, USA).

3.6.7. LC-MS/MS Analysis

The LC-MS/MS analysis was performed on an Agilent 1200 series (Agilent Technologies, Santa Clara, CA, USA) with an API 4000 linear ion trap triple quadruple mass spectrometer (AB Sciex, CA, USA). The chromatographic separation of the drug was carried out using a kinetex column (100 × 2.1 mm, 3 μm, Phenomenex, CA USA) with a SecurityGuard C18 guard column (4 mm × 2.0 mm i.d., Phenomenex). The flow rate was 300 µL/min with a mobile phase consisting of 0.1% formic acid in water (eluent A) and 0.1% formic acid in acetonitrile (eluent B) by linear gradient condition as follows: starting with 0 to 0.3 min, B: 5%; 0.3 to 2.5 min, B: 5 → 95%; 2.5 to 2.9 min, B: 95%; 2.9 to 3.0 min, B: 95 → 5%, and then re-equilibrated to initial conditions until 3 min. The total run time was 6 min and the injection volume of 5 μL.

3.6.8. In Vitro Screening Test

In vitro screening was performed as described previously [PMID: 33417443]. Briefly, TPH1 enzyme activity was measured with a commercially available kit (BPS Bioscience). Compounds were dissolved in DMSO (Sigma). Then, 10 μL of the compound was dispensed into a 96-well microplate, and 40 μL of the TPH1 enzyme solution (320 ng enzyme/reaction) was added. Then, 50 μL of the TPH1 reaction solution was added, and the microplate was sealed with aluminum foil. The microplate was immediately cooled to 4 °C, gently shaken, and incubated for 4 h. After a defined time, 10 μL of the TPH1 quench solution was added. Then, the fluorescence was measured with a Flexstation3 microplate reader at 300 nm for excitation and 360 nm for emission. All experiments were performed in triplicate.

3.6.9. In Vitro Efficacy Test

3T3-L1 cells (American Type Culture Collection, Manassas, VA, USA) were cultured in DMEM supplemented with 10% FCS and 100 mg mL−1 P/S in a humidified chamber under 5% atmospheric CO2 at 37 °C. Two days after reaching confluence, cells were further cultured in adipocyte differentiation medium (DMEM/10% FBS, P/S, 0.5 mM IBMX, 1 μg mL−1 insulin, and 1 μM dexamethasone) (day 0). After 2 days, the medium was changed to adipocyte maintenance medium (DMEM/10% FBS, 1 μg mL−1 insulin, P/S) (day 2), and the media was changed every 2 days from day 4 to 8. Cells were treated with compound 6 at 10 or 50 μM concentrations throughout adipocyte differentiation (day 0 thru day 8). On day 8, differentiated 3T3-L1 preadipocytes were harvested for analysis of gene expression.

3.6.10. In Vivo Efficacy Test

All animal study protocols were approved by the Institutional Animal Care and Use Committee at the Korea Advanced Institute of Science and Technology. Male C57BL/6J mice were purchased from SLC (Shizuoka, Japan) and acclimatized for a week. Mice were housed in a humidity and temperature-controlled environment under a 12 h light-dark cycle with ad libitum access to water and food. After acclimatization, male mice were fed a high-fat diet (HFD, 60% fat calories, Research diet; New Brunswick, NJ) and received daily treatment with TPH1 inhibitors. Intraperitoneal administration of PBS or 300 mg kg−1 pCPA (Sigma, St. Louis, MO, USA) was performed concomitant to the start of HFD feeding. Compound 18i was dissolved in 5% dimethyl sulfoxide (DMSO) (Sigma, St. Louis, MO, USA) and 25% β-cyclodextrin (Sigma, St. Louis, MO, USA). Vehicle or compound 18i (100 mg kg−1) was injected intraperitoneally in mice for 10 days after two weeks of HFD feeding. Both compounds 19 and 21 were prepared in 10% DMSO and 10% Kolliphor EL (Sigma, St. Louis, MO, USA). Vehicle and 100 mg kg−1 of compound 19 or compound 21 were administered daily by intraperitoneal injection or oral feeding, respectively, concomitant to the initiation of HFD feeding. For analyzing blood glucose concentration, glucose in blood collected from the tail vein was measured after fasting mice for 16 h using a Gluco Dr. Top glucometer (Allemedicus, Anyang, Korea). Body weight was measured daily and mice were sacrificed for harvesting adipose tissue and liver.

3.6.11. Histological Analysis and Immunohistochemistry

Tissue samples were fixed in 10% formalin, embedded in paraffin, and cut into 5 μm thick sections, followed by deparaffinization and rehydration for H&E staining and UCP1 immunohistochemistry. After heat-induced antigen retrieval with a citrate buffer (pH 6.0) for 10 min at 95°C, tissue sections were incubated with BLOXALL Blocking solution (Vector Laboratories, Burlingame, CA, USA) to block endogenous peroxidase, followed by incubation with 2% normal goat serum in phosphate-buffered saline (PBS) for 30 min at room temperature to block nonspecific binding. Sections were incubated overnight with the anti-UCP1 primary antibody diluted 1:200 in 2% normal goat serum in PBS (Abcam, Cambridge, UK) at 4 °C, followed by 30 min incubation with biotinylated anti-rabbit IgG secondary antibody diluted 1:400 in PBS for 30 min at room temperature. After incubation with Vectastain ABC-AP reagent (Vector Laboratories) for 30 min, peroxidase activity was visualized with 3,3′-diaminobenzidine substrates (Vector Laboratories). For measuring average adipocyte size, we analyzed three random fields of H&E stained micrographs per adipose tissue sample from two representative mice of each group using AdipoCount software [23].

3.6.12. Liver Oil-Red O Staining

Fresh liver tissues were flash frozen in O.C.T. compound (Sakura Finetek, Torrance, CA, USA) and 10 μm cryostat sections were collected on glass slides. Sections were allowed to air-dry for 1 h, rinsed with PBS for 5 min, and washed with distilled water for 5 min. After washing, sections were placed in 30% isopropanol for 5 min, 60% isopropanol for 5 min twice, and then stained in Oil Red O solution (0.5% Oil red O (Sigma, St. Louis, MO, USA) dissolved in 60% isopropanol) for 15 min. The stained sections were rinsed twice with distilled water, mounted, coverslipped, and documented using bright field microscopy.

3.6.13. RNA Isolation and Real-Time Quantitative PCR (RT-qPCR)

Total RNA was extracted from 3T3-L1 cells using Trizol reagent (Invitrogen, Carlsbad, CA, USA), purified using a RNeasy Lipid Tissue Mini kit (Qiagen, Hilden, Germany), and an aliquot (2 µg) of total RNA was reversed transcribed using the RevertAidTM First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. Real-time PCR was carried out using SYBR Green (Power SYBR Green PCR Master Mix, Thermo Fisher Scientific) with a ViiA 7 Real-Time PCR system (Applied Biosystems, Waltham, MA, USA). Mouse Rplp0 gene expression was used as the internal control to normalize gene expression values. Primer sequence information for RT-qPCR is shown in Supplementary Materials.

3.7. Docking Studies

The experimentally determined structure of TPH1 was downloaded from Protein Data Bank. The choice of three-dimensional structure [PDB access code 3HF8] is based on its high resolution, extensive literature, and our experience with the previous studies. Chain A was retrieved and preprocessed in a Discovery Studio Visualizer with a standard protocol to add missing atoms and residues. The structures of the synthesized compounds were manually drawn in ChemDraw and their 3D conformations were obtained from the Chem3D with the energy minimization protocol. The 3D conformations of TPH1 and synthesized compounds were preprocessed into ‘pdbqt’ files with AutoDockTools [11]. The docking studies were carried out with Lamarckian genetic algorithm implemented in AutoDock4.2 for 200 runs and 270,000 grid points of 0.325 grid spacing that covered the entire active site of TPH1 [11]. The detailed parameterization was described in our previous work [13]. The intermolecular interactions between the synthesized compounds and TPH1 were investigated with Discovery Studio Visualizer.

4. Conclusions

A series of oxyphenylalanine and heterocyclic phenylalanine derivatives were identified as TPH1 inhibitors. Among these derivatives, fused heterocyclic derivatives were identified as potent TPH1 inhibitors. Compound 18i with an IC50 value of 37 nM was the most active compound in vitro. Compound 18i showed good liver microsomal stability and did not significantly inhibit CYP and Herg. As a TPH1 inhibitor, this compound was able to interact with the peripheral system without penetrating the BBB. Compound 18i and its prodrugs, compounds 19 and 21, reduced body weight gain and decreased in vivo fat accumulation in mammals. Consequently, this compound as a therapeutic agent is a useful non-BBB permeable TPH1 inhibitor that can act on the peripheral system while preventing obesity and fatty liver disease.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/molecules27113417/s1, Experimental Section, Detailed General Procedure for the Synthesis of 6, 9a,b, 11a,b,15a,b, 18a–i, 19, 20, 21, 1H NMR and 13C NMR, HRMS, LC-MS, HPLC purity and melting point of compounds 6, 9a,b, 11a,b,15a,b, 18a–i, 19, 20, 21 and the sequence of primers for RT-qPCR.

Author Contributions

Methodology, J.-H.C. and J.S.S.; formal analysis, S.H.P., H.S.P., E.J.B., J.Y., S.P. (Saravanan Parameswaran), I.-K.L., H.K. and J.M.S.; investigation, S.P. (Sungmi Park), J.-H.J., M.A.B. and J.H.A.; data curation, K.-Y.P., K.-e.K. and M.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Ministry of Science, ICT and Future Planning (MSIP)/National Research Foundation of Korea (NRF) (2016M3A9B6902868, 2021R1A2C2008062). This work was supported by the Korea Institute of Energy Technology Evaluation and Planning (KETEP) grant funded by the Korea government (MOTIE) (20202020800330, Development and demonstration of energy efficient reaction separation purification process for fine chemical industry).

Institutional Review Board Statement

All animal study protocols were approved by the Institutional Animal Care and Use Committee at the Korea Advanced Institute of Science and Technology.

Acknowledgments

This work was supported by grants from the Ministry of Science, ICT and Future Planning (MSIP)/National Research Foundation of Korea (NRF) (2016M3A9B6902868, 2021R1A2C2008062). This work was supported by the Korea Institute of Energy Technology Evaluation and Planning (KETEP) grant funded by the Korean government (MOTIE) (20202020800330, Development and demonstration of energy efficient reaction separation purification process for fine chemical industry). This work was supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) and funded by the Ministry of Health and Welfare, Republic of Korea (HI16C1501).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BBB, Blood Brain Barriers; pCPA, para-chlorophenylalanine; TPH, tryptophan hydroxylase; 5-HT, 5-hydroxytryptamine; CCR2, CC chemokine receptor 2; CCL2, CC chemokine ligand 2; CCR5, CC chemokine receptor 5; TLC, thin layer chromatography; NaH, sodium hydride; DMF, Dimethylformamide; NaOH, sodium hydroxide; KOAc, potassium acetate; DMSO, Dimethyl sulfoxide; EtOAc, ethyl acetate; DCM, dichloromethane; THF, tetrahydrofuran; EtOH, ethanol.

References

  1. Berger, M.; Gray, J.A.; Roth, B.L. The Expanded Biology of Serotonin. Annu. Rev. Med. 2009, 60, 355–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Merens, W.; Willem Van der Does, A.J.; Spinhoven, P. The effects of serotonin manipulations on emotional information processing and mood. J. Affect. Disord. 2007, 103, 43–62. [Google Scholar] [CrossRef] [PubMed]
  3. Young, S.N.; Leyton, M. The role of serotonin in human mood and social interaction: Insight from altered tryptophan levels. Pharmacol. Biochem. Behav. 2002, 71, 857–865. [Google Scholar] [CrossRef]
  4. Monti, J.M. Serotonin control of sleep-wake behavior. Sleep Med. Rev. 2011, 15, 269–281. [Google Scholar] [CrossRef] [PubMed]
  5. Lam, D.D.; Garfield, A.S.; Marston, O.J.; Shaw, J.; Heisler, L.K. Brain serotonin system in the coordination of food intake and body weight. Pharmacol. Biochem. Behav. 2010, 97, 84–91. [Google Scholar] [CrossRef]
  6. Namkung, J.; Kim, H.; Park, S. Peripheral Serotonin: A New Player in Systemic Energy Homeostasis. Mol. Cells 2015, 38, 1023–1028. [Google Scholar]
  7. Oh, C.M.; Namkung, J.; Go, Y.; Shong, K.E.; Kim, K.; Kim, H.; Park, B.-Y.; Lee, H.W.; Jeon, Y.H.; Song, J.; et al. Regulation of systemic energy homeostasis by serotonin in adipose tissues. Nat. Commun. 2015, 6, 6794. [Google Scholar] [CrossRef]
  8. Available online: https://en.wikipedia.org/wiki/Fenclonine (accessed on 1 March 2022).
  9. Markham, A. Telotristat Ethyl: First Global Approval. Drugs 2017, 77, 793–798. [Google Scholar] [CrossRef]
  10. Goldberg, D.R.; De Lombaert, S.; Aiello, R.; Bourassa, P.; Barucci, N.; Zhang, Q.; Paralkar, V.; Stein, A.J.; Holt, M.; Valentine, J.; et al. Optimization of spirocyclic proline tryptophan hydroxylase-1 inhibitors. Bioorg. Med. Chem. Lett. 2017, 27, 413–419. [Google Scholar] [CrossRef]
  11. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. Autodock4 and AutoDockTools4: Automated docking with selective receptor flexiblity. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [Green Version]
  12. Cianchetta, G.; Stouch, T.; Yu, W.; Shi, Z.C.; Tari, L.W.; Swanson, R.V.; Hunter, M.J.; Hoffman, I.D.; Liu, Q. Mechanism of Inhibition of Novel Tryptophan Hydroxylase Inhibitors Revealed by Co-Crystal Structures and Kinetic Analysis. Curr. Chem. Genom. 2010, 4, 19–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bae, E.J.; Choi, W.G.; Pagire, H.S.; Pagire, S.H.; Parameswaran, S.; Choi, J.H.; Yoon, J.H.; Choi, W.I.; Lee, J.H.; Song, J.S.; et al. Peripheral Selective Oxadiazolylphenyl Alanine Derivatives as Tryptophan Hydroxylase 1 Inhibitors for Obesity and Fatty Liver Disease. J. Med. Chem. 2021, 64, 1037–1053. [Google Scholar] [CrossRef] [PubMed]
  14. Shi, Z.C.; Devasagayaraj, A.; Gu, K.; Jin, H.; Marinelli, B.; Samala, L.; Scott, S.; Stouch, T.; Tunoori, A.; Wang, Y.; et al. Modulation of Peripheral Serotonin Levels by Novel Tryptophan Hydroxylase Inhibitors for the Potential Treatment of Functional Gastrointestinal Disorders. J. Med. Chem. 2008, 51, 3684–3687. [Google Scholar] [CrossRef] [PubMed]
  15. Levin, V.A. Relationship of octanol/water partition coefficient and molecular weight to rat brain capillary permeability. J. Med. Chem. 1980, 23, 682–684. [Google Scholar] [CrossRef] [PubMed]
  16. Young, R.C.; Mitchell, R.C.; Brown, T.H.; Ganellin, C.R.; Griffiths, R.; Jones, M.; Rana, K.K.; Saunders, D.; Smith, I.R.; Sore, N.E.; et al. Development of a new physicochemical model for brain penetration and its application to the design of centrally acting H2 receptor histamine antagonists. J. Med. Chem. 1988, 31, 656–671. [Google Scholar] [CrossRef]
  17. Clark, D.E. Rapid calculation of polar molecular surface area and its application to the prediction of transport phenomena. 2. Prediction of blood–brain barrier penetration. J. Pharm. Sci. 1999, 88, 815–821. [Google Scholar] [CrossRef]
  18. Platts, J.A.; Abraham, M.H.; Zhao, Y.H.; Hersey, A.; Ijaz, L.; Butina, D. Correlation and prediction of a large blood–brain distribution data set—An LFER study. Eur. J. Med. Chem. 2001, 36, 719–730. [Google Scholar] [CrossRef]
  19. Seelig, A.; Gottschlich, R.; Devant, R.M. A method to determine the ability of drugs to diffuse through the blood-brain barrier. Proc. Natl. Acad. Sci. USA 1994, 91, 68–72. [Google Scholar] [CrossRef] [Green Version]
  20. Audus, K.L.; Ng, L.; Wang, W.; Borchardt, R.T. Brain Microvessel Endothelial Cell Culture Systems. In Models for Assessing Drug Absorption and Metabolism; Springer: Boston, MA, USA, 1996; pp. 239–258. [Google Scholar]
  21. Di, L.; Kerns, E.H.; Fan, K.; McConnell, O.J.; Carter, G.T. High throughput artificial membrane permeability assay for blood–brain barrier. Eur. J. Med. Chem. 2003, 38, 223–232. [Google Scholar] [CrossRef]
  22. Crane, J.D.; Palanivel, R.; Mottillo, E.P.; Bujak, A.L.; Wang, H.; Ford, R.J.; Collin, A.; Blümer, R.M.; Fullerton, M.D.; Yabut, J.M.; et al. Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nat. Med. 2015, 21, 166–172. [Google Scholar] [CrossRef] [Green Version]
  23. Zhi, X.; Wang, J.; Lu, P.; Shen, H.-B.; Ning, G. AdipoCount: A new software for automatic adipocyte counting. Front. Physiol. 2018, 9, 85. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The synthesis of serotonin (5-hydroxytryptamine [5-HT]).
Figure 1. The synthesis of serotonin (5-hydroxytryptamine [5-HT]).
Molecules 27 03417 g001
Scheme 1. Reagents and conditions: (a) SOCl₂, EtOH, reflux, 2 days, 63%; (b) (Boc)2O, TEA, MeOH, 0 °C to rt, 18 h, 96%; (c) Cs2CO3, ACN, 40 °C, 12 h, 87%; (d) NaOH, THF, H2O, rt, 12 h, 93%; (e) 4M HCl in dioxane, EtOAc, rt, 12 h, 50%; (f) 5a, b or 12, K2CO3, Pd(PPh3)4, H2O, dioxane, 100 °C, 2 h, 68%; (g) 8, PPh3, DIAD, THF, 0 °C to rt, 18 h, 98%; (h) Ammonia in IPA or aq.NaOH, dioxane, 50%; (i) (1) (CF3SO2)2O, DCM, pyridine, 0 °C-rt, 1 h; (2) Bis(pinacolato) diboron, PdCl2(dppf).DCM, KOAc, DMSO, 100 °C, 12 h, 84%.
Scheme 1. Reagents and conditions: (a) SOCl₂, EtOH, reflux, 2 days, 63%; (b) (Boc)2O, TEA, MeOH, 0 °C to rt, 18 h, 96%; (c) Cs2CO3, ACN, 40 °C, 12 h, 87%; (d) NaOH, THF, H2O, rt, 12 h, 93%; (e) 4M HCl in dioxane, EtOAc, rt, 12 h, 50%; (f) 5a, b or 12, K2CO3, Pd(PPh3)4, H2O, dioxane, 100 °C, 2 h, 68%; (g) 8, PPh3, DIAD, THF, 0 °C to rt, 18 h, 98%; (h) Ammonia in IPA or aq.NaOH, dioxane, 50%; (i) (1) (CF3SO2)2O, DCM, pyridine, 0 °C-rt, 1 h; (2) Bis(pinacolato) diboron, PdCl2(dppf).DCM, KOAc, DMSO, 100 °C, 12 h, 84%.
Molecules 27 03417 sch001
Scheme 2. Reagents and conditions: (a) 16c, 16di, NaH, DMF, 0 °C to rt, 96%; (b) 12 or 24ac, Pd(PPh3), K2CO3, dioxane, H2O, 90°C, 3 h, 70%; (c) NaOH, THF, H2O, rt; (d) 4M HCl in dioxane, EtOAc, rt, 12 h, 77%; (e) 4M HCl in dioxane, EtOAc, rt, 12 h; (f) aqueous ammonia; (g) hippuric acid, EtOH, reflux, 8 h, 95%; (h) 16a,b, dioxane, Cs2CO3, rt; (i) CF3Si(CH3)3, CsF, THF, 0 °C-rt, 6 h.
Scheme 2. Reagents and conditions: (a) 16c, 16di, NaH, DMF, 0 °C to rt, 96%; (b) 12 or 24ac, Pd(PPh3), K2CO3, dioxane, H2O, 90°C, 3 h, 70%; (c) NaOH, THF, H2O, rt; (d) 4M HCl in dioxane, EtOAc, rt, 12 h, 77%; (e) 4M HCl in dioxane, EtOAc, rt, 12 h; (f) aqueous ammonia; (g) hippuric acid, EtOH, reflux, 8 h, 95%; (h) 16a,b, dioxane, Cs2CO3, rt; (i) CF3Si(CH3)3, CsF, THF, 0 °C-rt, 6 h.
Molecules 27 03417 sch002
Scheme 3. Reagents and conditions: (a) Urea, 190 °C, 51%; (b) Br2, AcOH, 81%; (c) N,N-dimethylaniline, POCl3, 110 °C, 24 h, 87%; (d) DMF, NaH, 0 °C to rt, 46%; (e) 4-methoxybenzylamine, EtOH, TEA, MW 150 °C, 98%; (f) TFA, 120 °C, 12 h, 97%; (g) 12, Pd(PPh3), K2CO3, dioxane, H2O, 90 °C, 3 h, 86%; (h) (i) NaOH, THF, H2O, rt; (ii) 4M HCl in dioxane, EtOAc, rt, 12 h.
Scheme 3. Reagents and conditions: (a) Urea, 190 °C, 51%; (b) Br2, AcOH, 81%; (c) N,N-dimethylaniline, POCl3, 110 °C, 24 h, 87%; (d) DMF, NaH, 0 °C to rt, 46%; (e) 4-methoxybenzylamine, EtOH, TEA, MW 150 °C, 98%; (f) TFA, 120 °C, 12 h, 97%; (g) 12, Pd(PPh3), K2CO3, dioxane, H2O, 90 °C, 3 h, 86%; (h) (i) NaOH, THF, H2O, rt; (ii) 4M HCl in dioxane, EtOAc, rt, 12 h.
Molecules 27 03417 sch003
Figure 2. Binding analysis of compound 11b: Predicted binding mode of compound 11b with TPH1. TPH1 is shown as ribbons, while compound 11b is represented by sticks. The inset (left) shows the binding of compound 11b to the substrate-binding pocket of TPH1. The zoomed-in view (right) highlights the hydrophobic tail of compound 11b binding at the exterior of the active site of TPH1 with weak interactions. The entrance of the active site of TPH1 is not occupied with compound 11b which is required for the effective inhibition of TPH1.
Figure 2. Binding analysis of compound 11b: Predicted binding mode of compound 11b with TPH1. TPH1 is shown as ribbons, while compound 11b is represented by sticks. The inset (left) shows the binding of compound 11b to the substrate-binding pocket of TPH1. The zoomed-in view (right) highlights the hydrophobic tail of compound 11b binding at the exterior of the active site of TPH1 with weak interactions. The entrance of the active site of TPH1 is not occupied with compound 11b which is required for the effective inhibition of TPH1.
Molecules 27 03417 g002
Figure 3. Compound 18i protects against diet-induced obesity. (A) Lipogenic gene expression in 3T3-L1 cells treated with vehicle or 18i at the concentration of 10 μM or 50 μM. This experiment was repeated as biological triplicates with similar results (B) Representative haematoxylin and eosin (H&E) images of epididymal WAT (eWAT) and brown adipose tissue (BAT) from HFD-fed mice treated with vehicle or compound 18i-treated for 10 days (left, scale bar is 100 μm). Average adipocyte cross-section area of eWAT measured from H&E images (right). * p < 0.05, ** p < 0.01 and *** p < 0.001 vs. vehicle by Student’s t-test. Data are presented as mean ± s.e.m.
Figure 3. Compound 18i protects against diet-induced obesity. (A) Lipogenic gene expression in 3T3-L1 cells treated with vehicle or 18i at the concentration of 10 μM or 50 μM. This experiment was repeated as biological triplicates with similar results (B) Representative haematoxylin and eosin (H&E) images of epididymal WAT (eWAT) and brown adipose tissue (BAT) from HFD-fed mice treated with vehicle or compound 18i-treated for 10 days (left, scale bar is 100 μm). Average adipocyte cross-section area of eWAT measured from H&E images (right). * p < 0.05, ** p < 0.01 and *** p < 0.001 vs. vehicle by Student’s t-test. Data are presented as mean ± s.e.m.
Molecules 27 03417 g003
Figure 4. Compounds 19 and 21 show in vivo efficacy for obesity and fatty liver diseases. (A) Body weight of HFD-fed mice treated with vehicle (n = 6), 300 mg kg−1 pCPA (n = 3) or 100 mg kg−1 19 (n = 4) for 11 days. (B) Fasting blood glucose levels in HFD-fed mice after treatment of vehicle (n = 6), pCPA- (n = 3) or 19 (n = 4). (C) Representative H&E images of iWAT and eWAT from vehicle-, pCPA- or 19-treated mice fed a HFD (left, scale bar = 100 μm). The average adipocyte cross-section area of iWAT and eWAT measured from H&E images (right). (D) Immunohistochemistry of UCP1 protein in iWAT sections from HFD-fed mice treated with vehicle, pCPA, or compound 19 (scale bar = 100 μm) (E) Representative liver sections stained with H&E and Oil-red O (ORO) from HFD-fed mice treated with vehicle or compound 21 (scale bar = 100 μm). (F) Body and (G) liver weights after 6 weeks of vehicle or compound 21 treatment to HFD-fed mice. * p < 0.05, ** p < 0.01 and *** p < 0.001 vs. vehicle by Student’s t-test. Data are presented as mean ± s.e.m.
Figure 4. Compounds 19 and 21 show in vivo efficacy for obesity and fatty liver diseases. (A) Body weight of HFD-fed mice treated with vehicle (n = 6), 300 mg kg−1 pCPA (n = 3) or 100 mg kg−1 19 (n = 4) for 11 days. (B) Fasting blood glucose levels in HFD-fed mice after treatment of vehicle (n = 6), pCPA- (n = 3) or 19 (n = 4). (C) Representative H&E images of iWAT and eWAT from vehicle-, pCPA- or 19-treated mice fed a HFD (left, scale bar = 100 μm). The average adipocyte cross-section area of iWAT and eWAT measured from H&E images (right). (D) Immunohistochemistry of UCP1 protein in iWAT sections from HFD-fed mice treated with vehicle, pCPA, or compound 19 (scale bar = 100 μm) (E) Representative liver sections stained with H&E and Oil-red O (ORO) from HFD-fed mice treated with vehicle or compound 21 (scale bar = 100 μm). (F) Body and (G) liver weights after 6 weeks of vehicle or compound 21 treatment to HFD-fed mice. * p < 0.05, ** p < 0.01 and *** p < 0.001 vs. vehicle by Student’s t-test. Data are presented as mean ± s.e.m.
Molecules 27 03417 g004
Figure 5. The predicted binding mode of inhibitor 18i with TPH-1. TPH-1 is shown in ribbon representation with a partially transparent surface while the compound 18i is shown as sticks. The inset (left) shows the optimal binding of compound 18i at the active site of TPH-1 marked with the red box and the zoomed-in view (right) highlights the key interactions of hydrogen bonds, pi-sulfur, fluorine interactions, and hydrophobic interactions, which were depicted with green, yellow, cyan, and pink, respectively, according to their interaction type. The alkyl interactions were not shown for ease of presentation and clarity, which are explained in the main text. Figures were prepared with Discovery Studio Visualizer, Dassault Systèmes.
Figure 5. The predicted binding mode of inhibitor 18i with TPH-1. TPH-1 is shown in ribbon representation with a partially transparent surface while the compound 18i is shown as sticks. The inset (left) shows the optimal binding of compound 18i at the active site of TPH-1 marked with the red box and the zoomed-in view (right) highlights the key interactions of hydrogen bonds, pi-sulfur, fluorine interactions, and hydrophobic interactions, which were depicted with green, yellow, cyan, and pink, respectively, according to their interaction type. The alkyl interactions were not shown for ease of presentation and clarity, which are explained in the main text. Figures were prepared with Discovery Studio Visualizer, Dassault Systèmes.
Molecules 27 03417 g005
Table 1. In vitro test of thienopyrimidine derivatives.
Table 1. In vitro test of thienopyrimidine derivatives.
CompoundStructureTPH1 Inhibition
(% Inhibition at 100 µM)
pCPA Molecules 27 03417 i00125%
6 Molecules 27 03417 i00264%
9a Molecules 27 03417 i00345%
9b Molecules 27 03417 i00482%
11a Molecules 27 03417 i00558%
11b Molecules 27 03417 i00692%
15a Molecules 27 03417 i00749%
15b Molecules 27 03417 i00833%
18a Molecules 27 03417 i00990%
Table 2. In vitro test of thienopyrimidine derivatives.
Table 2. In vitro test of thienopyrimidine derivatives.
CompoundStructureTPH1 Inhibition
(% Inhibition at 1 µM)
IC50
(nM)
pCPA Molecules 27 03417 i010<5% (25% at 100 µM)
18a Molecules 27 03417 i01125% (90% at 100 µM)
18b Molecules 27 03417 i01210%
18c Molecules 27 03417 i0132.5% (10 µM)
18d Molecules 27 03417 i0141.2% (10 µM)
18e Molecules 27 03417 i01576%
18f Molecules 27 03417 i01641%
18g Molecules 27 03417 i01712%
18h Molecules 27 03417 i01878%263
18i Molecules 27 03417 i01995%37
Table 3. In vitro test of amino thienopyrimidine derivative and prodrugs of compound 18i.
Table 3. In vitro test of amino thienopyrimidine derivative and prodrugs of compound 18i.
CompoundStructureTPH1 Inhibition
(% Inhibition at 1 µM)
IC50
(nM)
LP533401 60%583
18i Molecules 27 03417 i02095%37
30 Molecules 27 03417 i02191%208
19 Molecules 27 03417 i02260%994
20 Molecules 27 03417 i02379%
21 Molecules 27 03417 i02492%
Table 4. PAMPA results.
Table 4. PAMPA results.
CompoundStructureBBB Permeability
Pe(10−6 cm/s)logPe
pCPA Molecules 27 03417 i0251.85 a−5.73
18i Molecules 27 03417 i0260.00−10.00
a High permeation predicted Pe (10−6 cm/s) > 4.0; low permeation predicted Pe (10−6 cm/s) < 2.0.
Table 5. Liver microsomal stability, CYP, and Herg inhibition of compound 18i.
Table 5. Liver microsomal stability, CYP, and Herg inhibition of compound 18i.
AssayResult
Liver microsomal stability (rat) a99% parent remained after 30 min incubation
Liver microsomal stability (human)99% parent remained after 30 min incubation
CYP inhibition1A2: 0.1% inhibition at 10 uM
2C19: 46.5% inhibition at 10 uM
2D6: 24.6% inhibition at 10 uM
3A4: 45.3% inhibition at 10 uM
Herg inhibition<1% inhibition at 10 uM
a Buspirone was used as a positive control (0.1% (rat) 4% (human) remained after 30 min incubation.
Table 6. BBB ratio and pharmacokinetic parameters of 18i in male rats.
Table 6. BBB ratio and pharmacokinetic parameters of 18i in male rats.
AssayResults
BBB penetrationPlasma: 0.5 h: 2266 ng/mL
3 h: 1439 ng/mL
Brain: 0.5 h: BQL a
3 h: BQL
In vivo PK (iv, 5 mpk)T1/2 (h): 1.85 ± 0.325
AUCt (μg·h/mL): 1.21 ± 0.058
AUC∞ (μg·h/mL): 1.22 ± 0.057
CL (L/h/kg): 4.09 ± 0.186
Vss (L/kg): 2.16 ± 1.62
(po, 5 mpk, prodrug 21)T1/2 (h): 7.95 ± 0.017
AUCt (μg·h/mL): 0.189 ± 0.009
AUC∞ (μg·h/mL): 0.216 ± 0.022
F = 16%
a below the quantification limit.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pagire, S.H.; Pagire, H.S.; Park, K.-Y.; Bae, E.J.; Kim, K.-e.; Kim, M.; Yoon, J.; Parameswaran, S.; Choi, J.-H.; Park, S.; et al. Identification of New Non-BBB Permeable Tryptophan Hydroxylase Inhibitors for Treating Obesity and Fatty Liver Disease. Molecules 2022, 27, 3417. https://doi.org/10.3390/molecules27113417

AMA Style

Pagire SH, Pagire HS, Park K-Y, Bae EJ, Kim K-e, Kim M, Yoon J, Parameswaran S, Choi J-H, Park S, et al. Identification of New Non-BBB Permeable Tryptophan Hydroxylase Inhibitors for Treating Obesity and Fatty Liver Disease. Molecules. 2022; 27(11):3417. https://doi.org/10.3390/molecules27113417

Chicago/Turabian Style

Pagire, Suvarna H., Haushabhau S. Pagire, Kun-Young Park, Eun Jung Bae, Kwang-eun Kim, Minhee Kim, Jihyeon Yoon, Saravanan Parameswaran, Jun-Ho Choi, Sungmi Park, and et al. 2022. "Identification of New Non-BBB Permeable Tryptophan Hydroxylase Inhibitors for Treating Obesity and Fatty Liver Disease" Molecules 27, no. 11: 3417. https://doi.org/10.3390/molecules27113417

Article Metrics

Back to TopTop