Next Article in Journal
Metabolomic Profiling of Iris palaestina via Molecular Networking and Its Anti-Diabetic Potential
Previous Article in Journal
Functional Fruit Snacks Enriched with Natural Sources of Fructooligosaccharides: Composition, Bioactive Compounds, Biological Activity, and Consumer Acceptance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Discovery of Novel Imidazothiazole-Based Hydroxamic Acid Derivatives as Potent Indoleamine 2,3-Dioxygenase 1 and Histone Deacetylase 6 Dual Inhibitors

1
Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China
2
Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China
3
Key Laboratory of Medical Biotechnology and Translational Medicine, School of Pharmacy, Guilin Medical University, Guilin 541199, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(12), 2508; https://doi.org/10.3390/molecules30122508
Submission received: 6 May 2025 / Revised: 4 June 2025 / Accepted: 5 June 2025 / Published: 7 June 2025

Abstract

:
In order to take advantage of both immunotherapeutic and epigenetic antitumor agents, a series of imidazothiazole-based hydroxamic acid derivatives were designed based on the pharmacophore fusion strategy and evaluated as potent IDO1 and HDAC6 dual inhibitors. Among these inhibitors, the most potent compound 3-(4-Bromophenyl)-N-{4-[(7-(hydroxyamino)-7-oxoheptyl)amino]phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10e) showed considerable IDO1 inhibitory activity and a good selectivity profile for HDAC6 over the other HDAC isoforms. The intracellular inhibition of HDAC6 by 10e was validated by Western blot analysis. Docking studies illustrated that the possible binding modes of compound 10e interacted with IDO1 and HDAC6. Moreover, compound 10e was found to arrest the cell cycle at the G2/M phase in HCT-116 cells. In particular, compound 10e also exhibited potent in vivo antitumor efficacy in CT26 tumor-bearing BALB/c mice models, with no significant toxicity. Collectively, this work provides a promising lead compound that serves as IDO1/HDAC6 dual inhibitor for the development of novel antitumor agents.

Graphical Abstract

1. Introduction

Immune checkpoint therapy has emerged as a groundbreaking advancement in cancer immunotherapy since the FDA approval of the CTLA-4 inhibitor ipilimumab and PD-1 blockers pembrolizumab and nivolumab [1]. Despite its success, the clinical benefits of anti-PD-1 monotherapy remain constrained by low response rates, immune-related adverse effects, and additional immunosuppressive factors in the tumor microenvironment (TME) [2,3]. To address these limitations, researchers are increasingly exploring combination approaches, such as integrating immunotherapy with chemotherapy or targeted therapy, some of which have shown promising clinical outcomes [4,5]. In addition, multitargeted agents—capable of modulating multiple pathways to enhance therapeutic efficacy—have garnered significant interest from both the industry and academia [6].
Indoleamine 2,3-dioxygenase 1 (IDO1), a heme-containing oxidoreductase that catalyzes tryptophan catabolism to kynurenine in the first rate-limiting step of the kynurenine pathway, plays an important role in tumor evasion of immune surveillance [7,8]. IDO1-induced immune tolerance is widely considered as one of the most critical mechanisms evolved by tumors to escape immune surveillance. It is constitutively overexpressed in many human tumors and host antigen-presenting cells, and has been correlated with different tumor progression parameters and poor prognosis [9]. Furthermore, IDO1 has been found to be involved in the suppression of CD8+ T effector cells and natural killer (NK) cells, increasing the activity of CD4+ regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs) [10,11]. Hence, IDO1 has been considered as an important target for immunotherapeutic intervention. At present, several small-molecule IDO1 inhibitors have entered clinical trials [12,13], such as Navoximod (NLG919), epacadostat, Linrodostat (BMS-986205), PF-06840003, and LY338191616 (Figure 1). In addition, imidazothiazole derivatives have been identified as potent IDO1 inhibitors [14,15]. However, preclinical and clinical studies indicated that IDO1 inhibitors only exhibited moderate antitumor activity when used as single agents [16].
Histone deacetylases (HDACs) are a family of important epigenetic enzymes that are responsible for controlling gene expression by modulating the acetylation status of histone and some nonhistone proteins and play a critical role in various cellular processes, such as transcription, the cell cycle, and cellular metabolism [17,18,19]. To date, five HDAC inhibitors (Figure 2)—Vorinostat, Romidepsin (FK 228), Panobinostat (LBH-589), Belinostat (PXD101), and Chidamide (CS055)—have been approved for the clinical treatment of hematological malignancies [20,21,22]. However, these known HDAC inhibitors are mainly pan-HDAC inhibitors, leading to undesirable side effects such as cardiotoxicity, and show little efficacy in the treatment of solid tumors as single agents [23,24]. HDAC6, predominantly a cytosolic member of the class IIb HDACs, is unique due to its ability to deacetylate a diverse set of nonhistone substrates such as α-tubulin and HSP90 [25]. HDAC6 is highly expressed in various cancer types, including malignant melanoma, bladder cancer, and lung cancer, and thus highlighting it as a potential therapeutic target for cancer treatment. Recent research studies have revealed that the selective inhibition of HDAC6 leads to the inhibition of tumorigenesis and metastasis, and to the increased sensitivity of tumors to other anticancer agents [26,27,28]. Furthermore, HDAC6 inhibitors have been reported to possess immunotherapeutic activity by decreasing programmed death-ligand 1 (PD-L1) expression by deactivating the STAT3 pathway [29,30]. Importantly, recent studies have demonstrated that a combination therapy composed of a selective HDAC6 inhibitor and a PD-1/PD-L1 inhibitor leads to significantly improved effects on tumor growth and antitumor immune response compared to a single therapy [31]. Therefore, the discovery of HDAC6 and immunotherapy-target dual inhibitors may provide a novel strategy for cancer treatment by taking advantage of both immunotherapeutic and epigenetic drugs.
Herein, we merged the common pharmacophores of HDAC inhibitors with the solvent exposure moieties of IDO1 inhibitors to design and synthesize a novel series of imidazothiazole-based hydroxamic acid derivatives as IDO1/HDAC6 dual inhibitors. Comprehensive evaluation identified the most potent inhibitor, 10e, having potential IDO1 inhibitory activity and good selectivity against HDAC6. Subsequent in vivo experiments indicated that compound 10e showed desirable in vivo antitumor efficacy in CT26 tumor-bearing mice.

2. Results

2.1. Rational Design of Novel IDO1 and HDAC6 Dual Inhibitors

The pharmacophore fusion strategy was carried out to design dual IDO1 and HDAC6 inhibitors. The imidazothiazole IDO1 inhibitor Amg-1 as well as the HDAC6 inhibitors Tubastatin A and ACY-1215 were used as the templates for the dual inhibitor design [15,32]. Generally, the classic pharmacophore model of HDAC inhibitors consists of a zinc-binding group (ZBG), a linker group, and a surface recognition cap group (Figure 3A) [33]. Among them, the cap group is a key point to gain subtype selectivity for HDAC inhibitors as it interacts with the surface of the wide basin around the entrance of the active site of the HDAC. On the other hand, according to the cocrystal structures of IDO1 and Amg-1 (Figure 3B), the nitrogen of the thiazolotriazole in Amg-1 directly coordinates with heme iron at the active site (pocket A) of IDO1, which has been identified as a crucial functional group for IDO1 inhibition [15]. The amide side chain of Amg-1, located at the expanded pocket B at the rim of the active site, could be modified without sacrificing IDO1 binding affinity. Thus, we fused the ZBG substructure into the IDO1 heme-binding imidazothiazole scaffold [14] via several different linkers (Figure 3C), and in this way, a series of novel IDO1/HDAC6 dual inhibitors were designed, synthesized, and biologically evaluated.

2.2. Chemistry

The detailed synthetic route to all the precursors for the target compounds (7a and 7b and 10a10k) is outlined in Scheme 1. Treatment of the commercially available 2-amino-4-(4-bromine phenyl) thiazole (1) with N,N-dimethylformamide dimethylacetal gave compound 2, which further reacted with ethyl bromoacetate to provide intermediate 3. Imidazothiazole compound 4 was formed in the presence of DBU, and the subsequent treatment of intermediate 4 with an alkaline solution yielded imidazothiazole carboxylic acid compound 5. The condensation of 5 with different amino-phenyl esters yielded ester 6, which was subsequently reacted with a hydroxylamine methanol solution to give the target compounds 7a and 7b. The condensation of 5 with N-Boc-p-phenylenediamine followed by Boc deprotection yielded compound 8, which reacted with commercially available bromo-alkanes under basic conditions to produce intermediate 9. Finally, target compounds 10a10k were prepared using conditions similar to those used for compounds 7a and 7b.

2.3. In Vitro IDO1/HDAC6 Enzyme Inhibition

Initially, all the target compounds were assayed for their inhibitory activity against IDO1 and HDAC6 with the IDO1 inhibitor epacadostat and the HDAC inhibitor SAHA as the positive controls. As shown in Table 1, compound 7a showed comparable HDAC6 inhibitory activity to SAHA, with 85.64% and 91.66% inhibition rates at 1 μM, respectively, but weaker IDO1 inhibitory activity than epacadostat. Generally, the linkers between the HDAC Zn-binding group and IDO1 heme-binding scaffold played an important role in enzyme inhibition. Compound 10a, with a two-carbon linker, showed limited inhibitory potency against both targets, while compound 10d, formed by extending the linker to a five-carbon chain, had significantly improved enzyme inhibition potency. The extension of the carbon chain resulted in increased activity against both targets. Specifically, compound 10e, with a linker of six carbons, exhibited the most robust potency against HDAC6, with an inhibition rate of 90.45% at 1 μM, and was slightly less potent than SAHA. On the other hand, the substituent at the R position also played a role in the enzyme inhibitory activities. In general, the compounds with a bromine substituent displayed better activities than the methyl-substituted compounds. Compound 7a, with a bromine substituent, was more potent than 7b. Similarly, the replacement of the bromine group in compound 10e with methyl (10k) resulted in decreased activity against both targets. Taken together, compound 10e, with a linker length of six atoms and a bromine substituent, demonstrated good inhibitory capacity against both IDO1 and HDAC6.

2.4. HDAC Isoform Selectivity of Compounds 7a, 10e, and 10k

To explore the inhibitory activity toward other HDAC isoforms, the selected compounds 7a, 10e, and 10k were further tested for their in vitro inhibitory activities against HDAC1, HDAC4, HDAC6, and HDAC11, with SAHA as the positive control. As shown in Table 2, all three compounds showed better activities against HDAC6 than HDAC1, HDAC4, and HDAC11. Compared with SAHA, all three selected compounds exhibited decreased inhibitory activities against HDAC1 and HDAC6, but showed a higher selectivity for HDAC6 than that of SAHA. Moreover, the HDAC6 inhibitory activity of compound 10e (IC50 = 58.23 nM) was better than that of 7a (IC50 = 84.44 nM). In particular, compound 10e had a slightly weaker inhibitory activity, but its selectivity toward HDAC6 was 7-fold higher than that of SAHA.

2.5. Binding Modes of Compound 10e with IDO1 and HDAC6

The above results confirm that compound 10e acted as a dual IDO1/HDAC6 inhibitor. Next, molecular docking analysis was performed to uncover the possible binding modes of compound 10e with IDO1 (PDB ID: 4PK5) [34] and HDAC6 (PDB ID: 6THV) [35], respectively, using SYBYL-X2.1 software. As shown in Figure 4A, compound 10e perfectly fitted into the binding interface formed by IDO1 (docking score: 11.97), and the central imidazothiazole scaffold of 10e directly bound to the heme iron and formed four hydrogen bonds with the porphyrin ring of heme, which contributed to its inhibitory activity against IDO1. The benzene ring on the imidazothiazole scaffold fitted into the hydrophobic pocket formed by the residues TYR126, VAL130, PHE163, PHE164, and LEU234. In addition, the π-π interactions were observed between the benzene ring and the key residues TYR126 and PHE164 of IDO1. Additionally, the carbonyl and amine moieties of the linker formed a hydrogen bond with the side chains of GLY262 and ARG231, respectively. In addition, the hydroxamic acid moiety also formed a stable hydrogen bond with key residue ARG231, enhancing its affinity. The binding mode of 10e with HDAC6 (docking score: 9.60) is displayed in Figure 4B. As expected, the hydroxamic acid of 10e coordinated with the catalytic zinc ion in the catalytic pocket and formed four hydrogen bonds with HIS614, HIS574, GLY582, and TYR745. Additionally, the amide part of the linker between the imidazothiazole scaffold and ZBG of compound 10e formed a hydrogen bond with ASP460 at the rim of the hHDAC6 binding pocket. Specifically, the terminal imidazothiazole scaffold of 10e, which also formed a hydrogen bond with ASP460, extended out of the binding site and was thus exposed to the solvent.

2.6. In Vitro Antiproliferative Activity of Selected Compounds

On the basis of the IDO1 and HDAC activities, we explored the proliferation inhibitory activities of the selected compounds against four solid tumor cells lines. The pan-HDAC SAHA was used as the positive control. The antiproliferative activities of selected compounds are shown in Table 3. As shown in Table 3, SAHA treatment generally resulted in strong antiproliferative effects, with IC50 values in the low micromolar range from 2.64 to 18.32 μM, whereas epacadostat was inactive against the four solid tumor cell lines. The data in Table 3 implied that all selected compounds and 5a possessed weak cytotoxicity against the tested cell lines, which was much weaker than that of SAHA. Additionally, compound 10e exhibited moderate antiproliferative activities against HCT-116 and SW480 cells, with IC50 values of 16.42 and 23.43 μM, respectively. It has been reported that IDO1 inhibitors do not destroy tumor cells directly. In addition, ricolinostat, an HDAC6 inhibitor with reduced class I HDAC inhibition, has low clinical activity when applied as monotherapy [36]. Noonepalle et al. showed that the selective HDAC6 inhibitor Suprastat was not cytotoxic up to 25 μM, while NextA began to induce cytotoxicity at a concentration of 10 μM [30]. Vögerl et al. also showed that selective HDAC6 inhibitors inhibited the proliferation of HUH7, MDA-MB-231, and T24 cells at concentrations of 10 and 100 μM after 72 h [37]. The possible reasons for the low cytotoxicity of 10e may be the alteration of key cancer-related pathways, such as immune checkpoints and unfolded protein response. It is worth noting that compounds 10e and 5a showed low toxicity to human normal renal cells compared with SAHA (IC50 = 26.60 ± 4.31 μM). These findings further emphasized that 10e was a selective HDAC6 inhibitor.

2.7. Inhibition of HDAC in HCT-116 Cells by Compound 10e

Since HDACs regulate the acetylation status of histone and nonhistone proteins in cells, the acetylation levels of histone H3 (a known substrate for HDACs 1, 2, and 3) and α-tubulin (a known substrate for HDAC6) are frequently used as markers of cellular HDAC activity. To assess the potency and isoform selectivity of 10e in cells, we performed a Western blot assay to investigate the in vitro inhibition of HDAC markers. HCT-116 cells were incubated with compound 10e (5−20 μM) for 24 h, and the levels of acetylation of histone H3 and α-tubulin were detected by the Western blot. As shown in Figure 5, compound 10e markedly increased Ac-α-tubulin levels in a concentration-dependent manner, and this feature was consistent with the signatory feature of the HDAC6 inhibitors. However, treatment with 10e led to a slight increase in the levels of Ac-histone H3 at high concentrations of 20 μM, which was consistent with a previous report [38]. These results demonstrated that compound 10e exhibited good HDAC6 potency and selectivity when tested in HCT-116 cells.

2.8. Cell Cycle Arrest

To assess the impact of compound 10e on the cell cycle, HCT-116 cells were treated with different concentrations of compound 10e (5 μM, 10 μM, and 20 μM) for 24 h, then stained with PI, and analyzed using flow cytometry. As displayed in Figure 6, compared with the control group (7.77%), the cell cycle analysis results revealed G2/M-phase cell cycle arrest, with increased cell population in the G2/M phase for 10e (17.39%, 31.89%, and 47.18%). These results suggested that compound 10e arrested the cell cycle of HCT-116 cells in the G2/M phase in a dose-dependent manner.

2.9. In Vivo Antitumor Effects

To further investigate the therapeutic capacity of compound 10e, its in vivo antitumor efficacy was evaluated in a CT26 tumor-bearing BALB/c mice model. After the average tumor volume reached 100 mm3 in each group, SAHA (150 mg/kg) and compound 10e (150 mg/kg) were administered intraperitoneally (ip) every 3 days for 21 consecutive days. As depicted in Figure 7A, compound 10e exhibited significant in vivo activity in inhibiting tumor growth compared with the vehicle group. At a dose of 150 mg/kg, compound 10e achieved a tumor growth inhibition (TGI) value of 60.2% (Figure 7B), which was more potent than SAHA (TGI = 38.7%). At the treatment end point, compound 10e treatment resulted in significantly smaller tumor volumes compared to the control group. The relative tumor volume growth rate (T/C) of 10e was 38.32% at 150 mg/kg, which was better than that of SAHA (58.97% at 150 mg/kg) (Figure 7C). Moreover, both 10e (150 mg/kg) and SAHA treatments displayed no obvious influence on the body weight of the mice in comparison with the vehicle group (Figure 7D). Additionally, the cellular morphology of the liver, spleen, and kidney in the H&E staining images revealed that no obvious histological differences were observed in the compound 10e-treated group (Figure 8), indicating its low toxicity toward normal organs. These results indicated that compound 10e effectively inhibited the growth of tumors with low general toxicity.

3. Materials and Methods

3.1. General Information

All raw materials, reagents, and solvents were analytical-grade and commercially available. Silica gel (200–300 mesh) was used for column chromatography. 1H NMR and 13C NMR spectra were recorded in DMSO-d6 on a 400 MHz (1H, 400 MHz; 13C, 101 MHz) Bruker spectrometer (AVANCE NEO 400M) or 600 MHz (1H, 600 MHz; 13C, 151 MHz) Bruker spectrometer (AVANCE NEO 600M) with TMS as an internal reference. High resolution mass spectra (HR-MS) were evaluated on an Agilent 1290–6545 UHPLC-QTOF mass spectrometers (Agilent Technologies, Santa Clara, CA, USA).

3.2. General Procedure for Preparation of Compounds 7a, 7b, and 10a10k

Equimolar ratios of 2-amino-4-(4-bromophenyl)thiazole or 2-amino-4-(4-tolyl)thiazole (6.00 g, 0.02 mol) with N,N-dimethylformamide dimethyl acetal (2.38 g, 0.02 mol) were refluxed with stirring in DMF (30 mL) at 70 °C for 3 h. Washings with water and ethyl acetate resulted in the concentration of compound 2. Compound 2 (5.9 g, 0.02 mol) was refluxed with ethyl bromoacetate (6.68 g, 0.04 mol) with stirring at 80 °C for 12 h. The resulting precipitate was filtered and washed with ethyl acetate to give the pale yellow bromide 3. Bromide 3 (5.37 g, 0.01 mol) was condensed and refluxed with DBU (0.03 mol) in DMF (30 mL) for 9 h and concentrated under reduced pressure to give compound 4. Compound 4 (5.37 g, 0.01 mol) was stirred in a mixture of tetrahydrofuran and methanol (30 mL) with 1N NaOH (22.5 mL) overnight, and the reaction was monitored by TLC. The reaction was concentrated under reduced pressure and acidified with 1N HCl to pH = 4; the precipitate was filtered and washed with water to obtain the white solid intermediate 5.
Compound 2a: HR-MS (m/z): calcd for C12H12BrN3S [M + H]+: 310.0013; found: 310.0033.
Compound 3a: HR-MS (m/z): calcd for C16H20BrN3O2S [M + H]+: 398.0538; found: 398.0375.
Compound 4a: HR-MS (m/z): calcd for C14H11BrN2O2S [M + H]+: 350.9803; found: 350.9818.
Intermediate 5a. 1H NMR (400 MHz, DMSO-d6) δ 12.57 (s, 1H), 7.92 (d, J = 1.2 Hz, 1H), 7.63 − 7.60 (m, 2H), 7.44 − 7.41 (m, 3H). 13C NMR (101 MHz, DMSO-d6) δ 159.21, 154.34, 142.17, 133.19, 130.66, 130.27, 130.12, 122.06, 120.77, 113.79. HR-MS (m/z) (ESI): calcd for C12H7BrN2O2S [M + H]+: 322.9490; found: 322.9492.
For the preparation of compounds 7a, 7b, and 10a10k, intermediate 5 (3.22 g, 0.01 mol) was mixed with EDCI (2.30 g, 0.012 mol), HOBT (1.62g, 0.012 mol), and Et3N (2.77 mL, 0.02 mol) in DCM solution at 0 °C, stirred for several minutes, and then ethyl 4-amino-benzoate or ethyl 4-aminomethyl-benzoate (1.98 g, 0.012 mol) was added and stirred overnight at room temperature. Silica gel chromatography (DCM/MeOH = 20: 1 v/v) was used to purify compound 6. Compound 6 was mixed with hydroxylamine hydrochloride methanol solution, stirred at 0 °C for 0.5 h, and then adjusted to pH = 7 with 1N HCl, filtered, and TCL purified to obtain compounds 7a and 7b. Compound 8 was synthesized in the same way as compound 6, and was first synthesized into compounds with Boc protecting groups and then deprotected with a 20% TFA/DCM solution to obtain compound 8. Compound 8 (4.12 g, 0.01 mol) was mixed with ethyl esters (0.01 mol) and DIPEA (4.85 mL, 0.03 mol) in DMF (15 mL) solution under stirring and reflux at 100 °C for 5–8 h to obtain compound 9. Compound 9 was reacted with hydroxylamine hydrochloride methanol solution at 0 °C and purified by TCL to obtain compounds 10a10k.
Compound 6a: HR-MS (m/z): calcd for C22H18BrN3O3S [M + H]+: 484.0330; found: 484.0325.
Compound 8a: HR-MS (m/z): calcd for C18H13BrN4OS [M + H]+: 413.0071; found: 413.0095.
Compound 9e: HR-MS (m/z): calcd for C27H29BrN4O3S [M + H]+: 569.1222; found: 569.1220.
3-(4-Bromophenyl)-N-[4-(hydroxycarbamoyl)benzyl]imidazo[2,1-b]thiazole-5-carboxamide (7a). Yield: 41.55%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.21 (s, 1H), 9.00 (t, J = 6.1 Hz, 1H), 7.81 − 7.79 (m, 1H), 7.76 − 7.73 (m, 2H), 7.49 − 7.46 (m, 2H), 7.40 (d, J = 1.1 Hz, 1H), 7.35 − 7.33 (m, 2H), 7.31 (d, J = 8.2 Hz, 2H), 4.27 (d, J = 6.0 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 158.35, 152.34, 142.69, 137.43, 132.67, 131.29, 130.80, 129.97, 129.19, 127.10, 126.88, 123.57, 121.91, 113.00, 41.88. HR-MS (m/z) (ESI): calcd for C20H15BrN4O3S [M + Na]+: 494.9926; found: 494.9918.
N-[4-(hydroxycarbamoyl)benzyl]-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (7b). Yield: 40.19%, as a white solid. 1H NMR (600 MHz, DMSO-d6) δ 11.22 (s, 1H), 9.04 (s, 1H), 8.95 (t, J = 6.1 Hz, 1H), 7.73 (d, J = 7.8 Hz, 3H), 7.30 (d, J = 8.0 Hz, 2H), 7.28 − 7.25 (m, 3H), 7.09 (d, J = 7.7 Hz, 2H), 4.24 (d, J = 6.1 Hz, 2H), 2.31 (s, 3H). 13C NMR (151 MHz, DMSO-d6) δ 164.01, 158.47, 152.13, 142.67, 138.15, 137.18, 133.85, 131.24, 128.49, 127.71, 127.21, 126.80, 123.71, 111.44, 41.96, 20.91. HR-MS (m/z) (ESI): calcd for C21H18N4O3S [M + H]+: 407.1180; found: 407.1171.
3-(4-Bromophenyl)-N-{4-[(3-(hydroxyamino)-3-oxopropyl)amino]phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10a). Yield: 33.69%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 9.92 (s, 1H), 8.79 (s, 1H), 7.84 (s, 1H), 7.57 (d, J = 8.4 Hz, 2H), 7.42 (s, 1H), 7.39 (d, J = 8.4 Hz, 2H), 7.13 (d, J = 8.8 Hz, 2H), 6.48 (d, J = 8.8 Hz, 2H), 5.46 (t, J = 5.9 Hz, 1H), 3.21 (d, J = 6.4 Hz, 2H), 2.22 (t, J = 7.1 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 167.73, 156.04, 152.09, 145.23, 137.51, 132.73, 131.00, 129.91, 129.21, 127.68, 124.17, 122.08, 121.77, 112.94, 111.84, 48.66, 32.34. HR-MS (m/z) (ESI): calcd for C21H18BrN5O3S [M + H]+: 500.0394; found: 500.0364.
3-(4-Bromophenyl)-N-{4-[(4-(hydroxyamino)-4-oxobutyl)amino)phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10b). Yield: 31.12%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H), 9.88 (s, 1H), 8.70 (s, 1H), 7.82 (s, 1H), 7.57 (d, J = 8.5 Hz, 2H), 7.42 (d, J = 1.1 Hz, 1H), 7.39 (d, J = 8.5 Hz, 2H), 7.11 (d, J = 8.9 Hz, 2H), 6.45 (d, J = 8.9 Hz, 2H), 5.46 (s, 1H), 2.94 (s, 2H), 2.04 (t, J = 7.4 Hz, 2H), 1.73 (t, J = 7.1 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.04, 155.99, 151.99, 145.63, 137.39, 132.71, 130.97, 129.87, 129.19, 127.38, 124.18, 122.04, 121.71, 112.85, 111.64, 42.67, 29.98, 24.80. HR-MS (m/z) (ESI): calcd for C22H20BrN5O3S [M + H]+: 516.0530; found: 516.0522.
3-(4-Bromophenyl)-N-{4-[(5-(hydroxyamino)-5-oxopentyl)amino)phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10c). Yield: 37.11%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.35 (s, 1H), 9.87 (s, 1H), 8.68 (s, 1H), 7.82 (s, 1H), 7.58 − 7.56 (m, 2H), 7.42 (d, J = 1.1 Hz, 1H), 7.41 − 7.38 (m, 2H), 7.10 (d, J = 8.9 Hz, 2H), 6.46 (d, J = 8.9 Hz, 2H), 5.42 (s, 1H), 2.95 (t, J = 6.8 Hz, 2H), 1.97 (t, J = 7.1 Hz, 2H), 1.56 (d, J = 7.8 Hz, 2H), 1.49 (t, J = 6.9 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.00, 155.97, 151.96, 145.76, 137.42, 132.69, 130.94, 129.87, 129.15, 127.24, 124.16, 121.99, 121.67, 112.86, 111.55, 42.74, 32.05, 28.23, 22.87. HR-MS (m/z) (ESI): calcd for C23H22BrN5O3S [M + H]+: 530.0686; found: 530.0682.
3-(4-Bromophenyl)-N-{4-[6-(hydroxyamino)-6-oxohexyl)amino]phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10d). Yield: 29.54%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.35 (s, 1H), 9.87 (s, 1H), 8.68 (d, J = 1.8 Hz, 1H), 7.82 (s, 1H), 7.58 − 7.56 (m, 2H), 7.42 (d, J = 1.3 Hz, 1H), 7.40 − 7.38 (m, 2H), 7.10 (d, J = 8.9 Hz, 2H), 6.46 − 6.44 (m, 2H), 5.41 (s, 1H), 2.93 (t, J = 7.1 Hz, 2H), 1.95 (t, J = 7.4 Hz, 2H), 1.51 (t, J = 7.6 Hz, 4H), 1.33 − 1.29 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.06, 155.97, 151.94, 145.79, 137.40, 132.68, 130.94, 129.89, 129.17, 127.28, 124.19, 122.00, 121.70, 112.83, 111.52, 43.02, 32.29, 28.47, 26.33, 25.05. HR-MS (m/z) (ESI): calcd for C24H24BrN5O3S [M + H]+: 544.0843; found: 544.0846.
3-(4-Bromophenyl)-N-{4-[(7-(hydroxyamino)-7-oxoheptyl)amino]phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10e). Yield: 30.97%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.36 (s, 1H), 9.87 (s, 1H), 8.68 (s, 1H), 7.82 (s, 1H), 7.56 (d, J = 8.1 Hz, 2H), 7.43 (s, 1H), 7.39 (d, J = 8.3 Hz, 2H), 7.10 (d, J = 8.5 Hz, 2H), 6.45 (d, J = 8.6 Hz, 2H), 5.37 (s, 1H), 2.93 (d, J = 5.4 Hz, 2H), 1.95 (t, J = 7.3 Hz, 2H), 1.50 (d, J = 6.4 Hz, 4H), 1.29 (dd, J = 24.0, 7.9 Hz, 4H). 13C NMR (101 MHz, DMSO-d6) δ 169.07, 155.97, 151.95, 145.81, 137.43, 132.68, 130.93, 129.87, 129.15, 127.25, 124.17, 121.99, 121.70, 112.85, 111.52, 43.09, 32.19, 28.62, 28.47, 26.44, 25.15. HR-MS (m/z) (ESI): calcd for C25H26BrN5O3S [M + H]+: 558.0999; found: 558.0971.
N-{4-[(2-(hydroxyamino)-2-oxoethyl)amino]phenyl}-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (10f). Yield: 34.86%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.59 (s, 1H), 9.90 (s, 1H), 8.83 (s, 1H), 7.77 (s, 1H), 7.34 − 7.32 (m, 2H), 7.31 (d, J = 1.1 Hz, 1H), 7.17 (d, J = 8.1 Hz, 2H), 7.09 (d, J = 8.9 Hz, 2H), 6.47 − 6.43 (m, 2H), 5.74 (t, J = 6.4 Hz, 1H), 3.53 (d, J = 6.3 Hz, 2H), 2.29 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 167.08, 156.14, 151.86, 144.91, 138.36, 137.25, 133.89, 128.65, 128.21, 127.65, 126.85, 124.27, 121.53, 112.05, 111.40, 44.69, 20.93. HR-MS (m/z) (ESI): calcd for C21H19N5O3S [M + Na]+: 444.1107; found: 444.1104.
N-{4-[(3-(hydroxyamino)-3-oxopropyl)amino]phenyl}-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (10g). Yield: 35.23%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.54 (s, 1H), 9.85 (s, 1H), 8.79 (s, 1H), 7.76 (s, 1H), 7.35 − 7.32 (m, 2H), 7.29 (d, J = 1.1 Hz, 1H), 7.17 (d, J = 8.3 Hz, 2H), 7.09 (d, J = 8.8 Hz, 2H), 6.46 (d, J = 8.9 Hz, 2H), 5.68 (t, J = 6.3 Hz, 1H), 4.08 (q, J = 5.3 Hz, 1H), 3.54 (d, J = 6.1 Hz, 1H), 3.17 (d, J = 5.3 Hz, 2H), 2.29 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 167.00, 156.11, 151.77, 144.86, 138.29, 137.15, 133.85, 128.57, 128.18, 127.60, 126.82, 124.25, 121.49, 112.02, 111.26, 48.56, 44.69, 20.85. HR-MS (m/z) (ESI): calcd for C22H21N5O3S [M + H]+: 436.1445; found: 436.1437.
N-{4-[(4-(hydroxyamino)-4-oxobutyl)amino]phenyl}-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (10h). Yield: 34.22%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.36 (s, 1H), 9.80 (s, 1H), 8.68 (s, 1H), 7.75 (s, 1H), 7.35 − 7.32 (m, 2H), 7.28 (d, J = 1.1 Hz, 1H), 7.16 (d, J = 8.1 Hz, 2H), 7.08 (d, J = 8.9 Hz, 2H), 6.45 − 6.42 (m, 2H), 5.41 (t, J = 5.6 Hz, 1H), 2.94 (d, J = 6.4 Hz, 2H), 2.28 (s, 3H), 2.04 (t, J = 7.4 Hz, 2H), 1.77 − 1.70 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 168.99, 156.06, 151.70, 145.53, 138.29, 137.05, 133.87, 128.59, 127.59, 127.46, 126.85, 124.33, 121.63, 111.52, 111.22, 42.66, 29.93, 24.78, 20.85. HR-MS (m/z) (ESI): calcd for C23H23N5O3S [M + H]+: 450.1602; found: 450.1593.
N-{4-[(5-(hydroxyamino)-5-oxopentyl)amino]phenyl}-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (10i). Yield: 28.13%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.35 (s, 1H), 9.80 (s, 1H), 8.66 (s, 1H), 7.75 (s, 1H), 7.35 − 7.32 (m, 2H), 7.28 (d, J = 1.0 Hz, 1H), 7.16 (d, J = 8.1 Hz, 2H), 7.07 (d, J = 8.9 Hz, 2H), 6.45 − 6.42 (m, 2H), 5.36 (t, J = 5.7 Hz, 1H), 2.94 (q, J = 6.6 Hz, 2H), 2.28 (s, 3H), 1.98 (t, J = 7.3 Hz, 2H), 1.61 − 1.54 (m, 2H), 1.52 − 1.45 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.00, 156.07, 151.70, 145.66, 138.29, 137.04, 133.87, 128.59, 127.60, 127.33, 126.85, 124.35, 121.66, 111.49, 111.23, 42.74, 32.04, 28.24, 22.84, 20.85. HR-MS (m/z) (ESI): calcd for C24H25N5O3S [M + H]+: 464.1758; found: 464.1745.
N-{4-[(6-(hydroxyamino)-6-oxohexyl)amino]phenyl}-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (10j). Yield: 38.31%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 9.81 (s, 1H), 8.66 (s, 1H), 7.75 (s, 1H), 7.33 (d, J = 8.1 Hz, 2H), 7.29 (d, J = 1.3 Hz, 1H), 7.16 (d, J = 8.0 Hz, 2H), 7.07 (d, J = 8.9 Hz, 2H), 6.44 − 6.42 (m, 2H), 5.35 (t, J = 5.7 Hz, 1H), 2.93 (d, J = 6.0 Hz, 2H), 2.28 (s, 3H), 1.95 (t, J = 7.4 Hz, 2H), 1.54 − 1.48 (m, 4H), 1.34 (d, J = 6.4 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 169.06, 156.06, 151.71, 145.72, 138.31, 137.08, 133.87, 128.62, 127.62, 127.34, 126.86, 124.35, 121.64, 111.47, 111.28, 43.01, 32.29, 28.47, 26.32, 25.03, 20.89. HR-MS (m/z) (ESI): calcd for C25H27N5O3S [M + H]+: 478.1915; found: 478.1900.
N-{4-[(7-(hydroxyamino)-7-oxoheptyl)amino]phenyl}-3-(p-tolyl)imidazo[2,1-b]thiazole-5-carboxamide (10k). Yield: 26.79%, as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 9.82 (s, 1H), 8.67 (s, 1H), 7.76 (s, 1H), 7.33 (d, J = 8.0 Hz, 2H), 7.29 (s, 1H), 7.16 (d, J = 8.1 Hz, 2H), 7.07 (d, J = 8.8 Hz, 2H), 6.43 (d, J = 8.9 Hz, 2H), 5.36 (t, J = 5.6 Hz, 1H), 2.93 (q, J = 6.7 Hz, 2H), 2.28 (s, 3H), 1.94 (t, J = 7.4 Hz, 2H), 1.53 − 1.46 (m, 4H), 1.38 − 1.26 (m, 4H). 13C NMR (101 MHz, DMSO-d6) δ 169.10, 156.06, 151.72, 145.74, 138.31, 137.09, 133.88, 128.63, 127.62, 127.33, 126.86, 124.36, 121.64, 111.47, 111.29, 43.08, 32.24, 28.62, 28.48, 26.43, 25.14, 20.89. HR-MS (m/z) (ESI): calcd for C26H29N5O3S [M + H]+: 492.2071; found: 492.2072.

3.3. Biological Assays

The biological experimental procedures, including the HDAC inhibitory assay, IDO1 inhibitory activity assay, wound healing assay, SPR assay, antiproliferative activity, molecular docking, cell cycle analysis, Western blot, and in vivo antitumor efficacy analyses were carried out according to our previous work [34,39,40,41] and are described in the Supplementary Information (SI).

4. Conclusions

In summary, a series of novel imidazothiazole hydroxamic acid derivatives were designed and synthesized as IDO1/HDAC6 dual inhibitors. Structure−activity relationship (SAR) exploration revealed several compounds as potent and selective IDO1/HDAC6 dual inhibitors, with IC50 values at the level of submicromolar concentrations. Particularly, the compound 3-(4-Bromophenyl)-N-{4-[(7-(hydroxyamino)-7-oxoheptyl)amino]phenyl}imidazo[2,1-b]thiazole-5-carboxamide (10e) was identified as a potent IDO1/HDAC6 dual inhibitor with superior HDAC6 inhibitory capacities to HDAC1, HDAC4, and HDAC11 as well. The HDAC6 selectivity profile of 10e was further supported by α-tubulin hyperacetylation. Docking studies illustrated the possible conformation of compound 10e binding to IDO1 and HDAC6. Dual inhibitor 10e displayed moderate antiproliferative activity against HCT-116 and SW480 cells, with IC50 values of 16.42 μM and 23.43 μM, respectively. Compound 10e was found to arrest the cell cycle at the G2/M phase in HCT-116 cells. Importantly, compound 10e exhibited good in vivo antitumor efficacy with no significant toxicity in a CT26 tumor-bearing BALB/c mice model. Consequently, these findings suggest that compound 10e is a potential IDO1/HDAC6 dual inhibitor with potent anticancer activity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules30122508/s1. Biological assays and Figures S1–S48. 1H-NMR, 13C-NMR, and HRMS spectra of all new compounds [15,35,42,43,44].

Author Contributions

X.-L.M. and R.-Z.H. conceived and designed the study; S.Z., Y.-F.W., and H.-R.L. performed the experiments; X.-Q.Y. performed the database search; Y.Z. provided overall supervision and guidance. All the authors analyzed data, discussed the results, and contributed to writing the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Guangxi Natural Science Foundation (Nos. 2023GXNSFBA026303), Guangxi Science and Technology Major Program (No. Guike AA24263012), Guangxi Key Laboratory of Drug Discovery and Optimization (No. 23-026-02), State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (No. CMEMR2023-B17), Guilin Scientific Research and Technology Development Plan Project (No. 20220129-2), and the College Students’ Innovative Entrepreneurial Training Plan Program (No. S202410601136).

Institutional Review Board Statement

All the procedures were approved by the Animal Care and Use Committee of Guilin Medical University (No. GLMC202303200).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef] [PubMed]
  2. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  3. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef] [PubMed]
  4. Kirchhammer, N.; Trefny, M.P.; der Maur, P.A.; Läubli, H.; Zippelius, A. Combination cancer immunotherapies: Emerging treatment strategies adapted to the tumor microenvironment. Sci. Transl. Med. 2022, 14, eabo3605. [Google Scholar] [CrossRef] [PubMed]
  5. Hughes, P.E.; Caenepee, S.; Wu, L.C. Targeted Therapy and Checkpoint Immunotherapy Combinations for the Treatment of Cancer. Trends Immunol. 2016, 37, 462–476. [Google Scholar] [CrossRef]
  6. Li, X.; Li, X.; Liu, F.; Li, S.; Shi, D. Rational Multitargeted Drug Design Strategy from the Perspective of a Medicinal Chemist. J. Med. Chem. 2021, 64, 10581–10605. [Google Scholar] [CrossRef]
  7. Ricciuti, B.; Leonardi, G.C.; Puccetti, P.; Fallarino, F.; Bianconi, V.; Sahebkar, A.; Baglivo, S.; Chiari, R.; Pirro, M. Targeting indoleamine-2,3-dioxygenase in cancer: Scientific rationale and clinical evidence. Pharmacol. Therapeut. 2019, 196, 105–116. [Google Scholar] [CrossRef]
  8. Li, F.; Zhang, R.; Li, S.; Liu, J. IDO1: An important immunotherapy target in cancer treatment. Int. Immunopharmacol. 2017, 47, 70–77. [Google Scholar] [CrossRef]
  9. Weng, T.; Qiu, X.; Wang, J.; Li, Z.; Bian, J. Recent discovery of indoleamine-2,3-dioxygenase 1 inhibitors targeting cancer immunotherapy. Eur. J. Med. Chem. 2018, 143, 656–669. [Google Scholar] [CrossRef]
  10. Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef]
  11. Prendergast, G.C.; Mondal, A.; Dey, S.; Laury-Kleintop, L.D.; Muller, A.J. Inflammatory Reprogramming with IDO1 Inhibitors: Turning Immunologically Unresponsive ‘Cold’ Tumors ‘Hot’. Trends. Cancer 2018, 4, 38–58. [Google Scholar] [CrossRef] [PubMed]
  12. NayakKapoor, A.; Hao, Z.; Sadek, R.; Dobbins, R.; Marshall, L.; Vahanian, N.N.; Ramsey, W.J.; Kennedy, E.; Mautino, M.R.; Link, C.J.; et al. Phase Ia study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) in patients with recurrent advanced solid tumors. Cancer Immunol. 2018, 6, 1–12. [Google Scholar]
  13. Feng, X.; Liao, D.; Liu, D.; Ping, A.; Li, Z.; Bian, J. Development of Indoleamine 2,3-Dioxygenase 1 Inhibitors for Cancer Therapy and Beyond: A Recent Perspective. J. Med. Chem. 2020, 63, 15115–15139. [Google Scholar] [CrossRef] [PubMed]
  14. Peng, Y.-H.; Liao, F.-Y.; Tseng, C.-T.; Kuppusamy, R.; Li, A.-S.; Chen, C.-H.; Fan, Y.-S.; Wang, S.-Y.; Wu, M.-H.; Hsueh, C.-C.; et al. Unique sulfur–aromatic interactions contribute to the binding of potent imidazothiazole indoleamine 2,3-dioxygenase inhibitors. J. Med. Chem. 2020, 63, 1642–1659. [Google Scholar] [CrossRef]
  15. Tojo, S.; Kohno, T.; Tanaka, T.; Kamioka, S.; Ota, Y.; Ishii, T.; Kamimoto, K.; Asano, S.; Isobe, Y. Crystal Structures and Structure–Activity Relationships of Imidazothiazole Derivatives as IDO1 Inhibitors. ACS Med. Chem. Lett. 2014, 5, 1119–1123. [Google Scholar] [CrossRef]
  16. Zhang, Y.; Hu, Z.; Zhang, J.; Ren, C.; Wang, Y. Dual-target inhibitors of indoleamine 2, 3 dioxygenase 1 (Ido1): A promising direction in cancer immunotherapy. Eur. J. Med. Chem. 2022, 238, 114524. [Google Scholar] [CrossRef]
  17. Ru, J.; Wang, Y.; Li, Z.; Wang, J.; Ren, C.; Zhang, J. Technologies of targeting histone deacetylase in drug discovery: Current progress and emerging prospects. Eur. J. Med. Chem. 2023, 261, 115800. [Google Scholar] [CrossRef]
  18. Pan, Y.; Hou, H.; Zhou, B.; Gao, J.; Gao, F. Hydroxamic acid hybrids: Histone deacetylase inhibitors with anticancer therapeutic potency. Eur. J. Med. Chem. 2023, 262, 115879. [Google Scholar] [CrossRef]
  19. Liang, T.; Wang, F.; Elhassan, R.M.; Cheng, Y.; Tang, X.; Chen, W.; Fang, H.; Hou, X. Targeting histone deacetylases for cancer therapy: Trends and challenges. Acta Pharm. Sin. B 2023, 13, 2425–2463. [Google Scholar] [CrossRef]
  20. Zang, J.; Liang, X.; Huang, Y.; Jia, Y.; Li, X.; Xu, W.; Chou, C.J.; Zhang, Y. Discovery of Novel Pazopanib-Based HDAC and VEGFR Dual Inhibitors Targeting Cancer Epigenetics and Angiogenesis Simultaneously. J. Med. Chem. 2018, 61, 5304–5322. [Google Scholar] [CrossRef]
  21. Suraweera, A.; O’Byrne, K.J.; Richard, D.J. Combination Therapy with Histone Deacetylase Inhibitors (HDACi) for the Treatment of Cancer: Achieving the Full Therapeutic Potential of HDACi. Front. Oncol. 2018, 8, 92. [Google Scholar] [CrossRef] [PubMed]
  22. Li, Y.; Wang, F.; Chen, X.; Wang, J.; Zhao, Y.; Li, Y.; He, B. Zinc-dependent deacetylase (HDAC) inhibitors with different zinc binding groups. Curr. Top. Med. Chem. 2019, 19, 223–241. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, T.; Wan, Y.; Xiao, Y.; Xia, C.; Duan, G. Dual-Target Inhibitors Based on HDACs: Novel Antitumor Agents for Cancer Therapy. J. Med. Chem. 2020, 63, 8977–9002. [Google Scholar] [CrossRef]
  24. Peng, X.; Sun, Z.; Kuang, P.; Chen, J. Recent progress on HDAC inhibitors with dual targeting capabilities for cancer treatment. Eur. J. Med. Chem. 2020, 208, 112831. [Google Scholar] [CrossRef]
  25. Zhang, X.-H.; Ma, Q.; Wu, H.-P.; Khamis, M.Y.; Li, Y.-H.; Ma, L.-Y.; Liu, H.-M. A Review of Progress in Histone Deacetylase 6 Inhibitors Research: Structural Specificity and Functional Diversity. J. Med. Chem. 2021, 64, 1362–1391. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, X.; Wang, J.; Zhao, P.; Dang, B.; Liang, T.; Steimbach, R.R.; Miller, A.K.; Liu, J.; Wang, X.; Zhang, T.; et al. Tetrahydro-β-carboline derivatives as potent histone deacetylase 6 inhibitors with broad-spectrum antiproliferative activity. Eur. J. Med. Chem. 2023, 260, 115776. [Google Scholar] [CrossRef]
  27. Wu, H.; Yang, K.; Zhang, Z.; Leisten, E.D.; Li, Z.; Xie, H.; Liu, J.; Smith, K.A.; Novakova, Z.; Barinka, C.; et al. Development of Multifunctional Histone Deacetylase 6 Degraders with Potent Antimyeloma Activity. J. Med. Chem. 2019, 62, 7042–7057. [Google Scholar] [CrossRef]
  28. Zeng, H.; Qu, J.; Jin, N.; Xu, J.; Lin, C.; Chen, Y.; Yang, X.; He, X.; Tang, S.; Lan, X.; et al. Feedback Activation of Leukemia Inhibitory Factor Receptor Limits Response to Histone Deacetylase Inhibitors in Breast Cancer. Cancer Cell 2016, 30, 459–473. [Google Scholar] [CrossRef]
  29. Chen, X.; Pan, X.; Zhang, W.; Guo, H.; Cheng, S.; He, Q.; Yang, B.; Ding, L. Epigenetic strategies synergize with PD-L1/PD-1 targeted cancer immunotherapies to enhance antitumor responses. Acta Pharm. Sin. B 2020, 10, 723–733. [Google Scholar] [CrossRef]
  30. Noonepalle, S.; Shen, S.; Ptáček, J.; Tavares, M.T.; Zhang, G.; Stránský, J.; Pavlíček, J.; Ferreira, G.M.; Hadley, M.; Pelaez, G.; et al. Rational Design of Suprastat: A Novel Selective Histone Deacetylase 6 Inhibitor with the Ability to Potentiate Immunotherapy in Melanoma Models. J. Med. Chem. 2020, 63, 10246–10262. [Google Scholar] [CrossRef]
  31. Peng, X.; Li, L.; Chen, J.; Ren, Y.; Liu, J.; Yu, Z.; Cao, H.; Chen, J. Discovery of Novel Histone Deacetylase 6 (HDAC6) Inhibitors with Enhanced Antitumor Immunity of Anti-PD-L1 Immunotherapy in Melanoma. J. Med. Chem. 2022, 65, 2434–2457. [Google Scholar] [CrossRef] [PubMed]
  32. Miyake, Y.; Keusch, J.J.; Wang, L.; Saito, M.; Hess, D.; Wang, X.; Melancon, B.J.; Helquist, P.; Gut, H.; Matthias, P. Structural insights into HDAC6 tubulin deacetylation and its selective inhibition. Nat. Chem. Biol. 2016, 12, 748–754. [Google Scholar] [CrossRef] [PubMed]
  33. Jiang, Q.; Tang, Y.; Hu, Q.; Wang, B.; Ruan, X.; Zhou, Q. Discovery of novel itaconimide-based derivatives as potent HDAC inhibitors for the efficient treatment of prostate cancer. Eur. J. Med. Chem. 2024, 269, 116315. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, K.; Song, L.-H.; Liang, Q.-L.; Zhang, Y.; Ma, X.-L.; Wang, Q.; Zhang, H.-Y.; Jiang, C.-N.; Wei, J.-H.; Huang, R.-Z. Discovery of novel sulfonamide chromone-oxime derivatives as potent indoleamine 2,3-dioxygenase 1 inhibitors. Eur. J. Med. Chem. 2023, 254, 115349. [Google Scholar] [CrossRef]
  35. Shen, S.; Svoboda, M.; Zhang, G.; Cavasin, M.A.; Motlova, L.; McKinsey, T.A.; Eubanks, J.H.; Barinka, C.; Kozikowski, A.P. Structural and in Vivo Characterization of Tubastatin A, a Widely Used Histone Deacetylase 6 Inhibitor. ACS Med. Chem. Lett. 2020, 11, 706–712. [Google Scholar] [CrossRef]
  36. Yee, A.J.; Bensinger, W.I.; Supko, J.G.; Voorhees, P.M.; Berdeja, J.G.; Richardson, P.G.; Libby, E.N.; Wallace, E.E.; Birrer, N.E.; Burke, J.N.; et al. Ricolinostat plus lenalidomide, and dexamethasone in relapsed or refractory multiple myeloma: A multicentre phase Ib trial. Lancet Oncol. 2016, 17, 1569–1578. [Google Scholar] [CrossRef] [PubMed]
  37. Vögerl, K.; Ong, N.; Senger, J.; Herp, D.; Schmidtkunz, K.; Marek, M.; Müller, M.; Bartel, K.; Shaik, T.B.; Porter, N.J.; et al. Synthesis and Biological Investigation of Phenothiazine-Based Benzhydroxamic Acids as Selective Histone Deacetylase 6 Inhibitors. J. Med. Chem. 2019, 62, 1138–1166. [Google Scholar] [CrossRef]
  38. Yang, Z.; Wang, T.; Wang, F.; Niu, T.; Liu, Z.; Chen, X.; Long, C.; Tang, M.; Cao, D.; Wang, X.; et al. Discovery of selective histone deacetylase 6 inhibitors using the quinazoline as the cap for the treatment of cancer. J. Med. Chem. 2016, 59, 1455–1470. [Google Scholar] [CrossRef]
  39. Liang, S.-M.; Liang, G.-B.; Wang, H.-L.; Jiang, H.; Ma, X.-L.; Wei, J.-H.; Huang, R.-Z.; Zhang, Y. Discovery of 4-(N-dithiobenzyl piperazine)-1,8-naphthalimide as a potent multi-target antitumor agent with good efficacy, limited toxicity, and low resistance. Eur. J. Med. Chem. 2024, 263, 115937. [Google Scholar] [CrossRef]
  40. Liang, Q.; Zhang, S.; Liu, J.; Zhou, X.; Ariffin, N.S.; Wei, J.; Shi, C.; Ma, X.; Zhang, Y.; Huang, R. Discovery of novel 1,8-naphthalimide piperazinamide based benzenesulfonamides derivatives as potent carbonic anhydrase IX inhibitors and ferroptosis inducers for the treatment of triple-negative breast cancer. Bioorg. Chem. 2024, 150, 107596. [Google Scholar] [CrossRef]
  41. Liu, S.-Q.; Mao, Z.-C.; Xu, Y.-L.; Chen, X.-M.; Wang, H.-L.; Wang, Q.; Wei, J.-H.; Huang, R.-Z.; Zhang, Y. Discovery of two biotin-PEG4-diarylidenyl piperidone prodrugs as potent antitumor agents with good efficacy, limited toxicity, and low resistance. Bioorg. Chem. 2023, 131, 106323. [Google Scholar] [CrossRef] [PubMed]
  42. Huang, R.; Jing, X.; Huang, X.; Pan, Y.; Fang, Y.; Liang, G.; Liao, Z.; Wang, H.; Chen, Z.; Zhang, Y. Bifunctional naphthoquinone aromatic amide-oxime derivatives exert combined immunotherapeutic and antitumor effects through simultaneous targeting of indoleamine-2,3-dioxygenase and signal transducer and activator of transcription 3. J. Med. Chem. 2020, 63, 1544–1563. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, X.; Wang, M.; You, Q.; Kong, J.; Zhang, H.; Yu, C.; Wang, Y.; Wang, H.; Huang, R. Synthesis, mechanisms of action, and toxicity of novel aminophosphonates derivatives conjugated irinotecan in vitro and in vivo as potent antitumor agents. Eur. J. Med. Chem. 2020, 189, 112067. [Google Scholar] [CrossRef] [PubMed]
  44. Liang, G.-B.; Wei, J.-H.; Jiang, H.; Huang, R.-Z.; Qin, J.-T.; Wang, H.-L.; Wang, H.-S.; Zhang, Y. Design, synthesis and antitumor evaluation of new 1,8-naphthalimide derivatives targeting nuclear DNA. Eur. J. Med. Chem. 2021, 210, 112951. [Google Scholar] [CrossRef]
Figure 1. Structures of candidates targeting IDO1 in clinical trial.
Figure 1. Structures of candidates targeting IDO1 in clinical trial.
Molecules 30 02508 g001
Figure 2. Structures and pharmacophores of representative HDAC inhibitors.
Figure 2. Structures and pharmacophores of representative HDAC inhibitors.
Molecules 30 02508 g002
Figure 3. The design strategy of the IDO1/HDAC6 dual inhibitors. (A) The binding model of Amg-1 bound to IDO1 (PDB code 4PK5). The proteins are shown as 3D models. The carbon atoms of Amg-1 are shown in pink, and heme in yellow. (B) The binding model of Tubastatin A bound to HDAC6 (PDB code 6THV). The Tubastatin A is shown as sticks, with carbon atoms in cyan. (C) The pharmacophore fusion strategy for the design of the dual inhibitors.
Figure 3. The design strategy of the IDO1/HDAC6 dual inhibitors. (A) The binding model of Amg-1 bound to IDO1 (PDB code 4PK5). The proteins are shown as 3D models. The carbon atoms of Amg-1 are shown in pink, and heme in yellow. (B) The binding model of Tubastatin A bound to HDAC6 (PDB code 6THV). The Tubastatin A is shown as sticks, with carbon atoms in cyan. (C) The pharmacophore fusion strategy for the design of the dual inhibitors.
Molecules 30 02508 g003
Scheme 1. Synthetic routes of compounds 7a, 7b, and 10a10k. Reagents and conditions: (a) N,N-Dimethylformamide dimethyl acetal, DMF, 70 °C, 3 h; (b) Ethyl 2-bromoacetate, 80 °C, 12 h; (c) DBU, DMF, 80 °C, 9 h; (d) NaOH, THF/MeOH, rt, 12 h; (e) EDCI, HOBT, Et3N, CH2Cl2, 0 °C, rt, 12 h; (f) NH2OH·HCl, MeOH, 0 °C, 0.5 h; (g) EDCI, HOBT, Et3N, CH2Cl2, 0 °C, rt, 12 h; (h) DIPEA, DMF, 100 °C, 5–8 h; (i) NH2OH·HCl, MeOH, 0 °C, 0.5 h.
Scheme 1. Synthetic routes of compounds 7a, 7b, and 10a10k. Reagents and conditions: (a) N,N-Dimethylformamide dimethyl acetal, DMF, 70 °C, 3 h; (b) Ethyl 2-bromoacetate, 80 °C, 12 h; (c) DBU, DMF, 80 °C, 9 h; (d) NaOH, THF/MeOH, rt, 12 h; (e) EDCI, HOBT, Et3N, CH2Cl2, 0 °C, rt, 12 h; (f) NH2OH·HCl, MeOH, 0 °C, 0.5 h; (g) EDCI, HOBT, Et3N, CH2Cl2, 0 °C, rt, 12 h; (h) DIPEA, DMF, 100 °C, 5–8 h; (i) NH2OH·HCl, MeOH, 0 °C, 0.5 h.
Molecules 30 02508 sch001
Figure 4. Proposed binding modes of compound 10e (yellow) to IDO1 and HDAC6. (A) Compound 10e docked into IDO1 (PDB ID: 4PK5). (B) Compound 10e docked into HDAC6 (PDB ID: 6THV).
Figure 4. Proposed binding modes of compound 10e (yellow) to IDO1 and HDAC6. (A) Compound 10e docked into IDO1 (PDB ID: 4PK5). (B) Compound 10e docked into HDAC6 (PDB ID: 6THV).
Molecules 30 02508 g004
Figure 5. Western blots of Ac-α-tubulin and Ac-histone H3 after treatment of HCT-116 cells with compound 10e. GAPDH was loading control.
Figure 5. Western blots of Ac-α-tubulin and Ac-histone H3 after treatment of HCT-116 cells with compound 10e. GAPDH was loading control.
Molecules 30 02508 g005
Figure 6. The cell cycle arrest effects of compound 10e in HCT-116 cells. The cells were treated with 5, 10, and 20 μM of 10e for 24 h, respectively, then stained with PI, and assessed by flow cytometry.
Figure 6. The cell cycle arrest effects of compound 10e in HCT-116 cells. The cells were treated with 5, 10, and 20 μM of 10e for 24 h, respectively, then stained with PI, and assessed by flow cytometry.
Molecules 30 02508 g006
Figure 7. The in vivo antitumor activities of SAHA and compound 10e against CT26 tumors in BALB/c mice. (A) Images of the harvested tumors from mice after the administration of SAHA (150 mg/kg) and 10e (150 mg/kg) every 3 days for 3 weeks. (B) The tumor weights of the excised tumors of each group. (C) The tumor volume of the mice in each group during the observation period. (D) The body weight of the mice in each group at the end of the observation period. These data are presented as means ± SD. p < 0.05 (versus the vehicle control group).
Figure 7. The in vivo antitumor activities of SAHA and compound 10e against CT26 tumors in BALB/c mice. (A) Images of the harvested tumors from mice after the administration of SAHA (150 mg/kg) and 10e (150 mg/kg) every 3 days for 3 weeks. (B) The tumor weights of the excised tumors of each group. (C) The tumor volume of the mice in each group during the observation period. (D) The body weight of the mice in each group at the end of the observation period. These data are presented as means ± SD. p < 0.05 (versus the vehicle control group).
Molecules 30 02508 g007
Figure 8. Histopathological changes in major organs dissected from tumor-bearing mice after 10e treatment by H&E staining.
Figure 8. Histopathological changes in major organs dissected from tumor-bearing mice after 10e treatment by H&E staining.
Molecules 30 02508 g008
Table 1. In vitro enzyme inhibitory activity of compounds 7a, 7b, and 10a10k against IDO1 and HDAC6.
Table 1. In vitro enzyme inhibitory activity of compounds 7a, 7b, and 10a10k against IDO1 and HDAC6.
Molecules 30 02508 i001
Compd.RnIDO1
IC50 (μM) a
HDAC6 Inhibition Rate at 1 μM
7aBr10.53 ± 0.1185.64%
7bMe10.96 ± 0.1347.95%
10aBr21.87 ± 0.0622.44%
10bBr31.06 ± 0.0225.24%
10cBr40.34 ± 0.0919.37%
10dBr50.12 ± 0.0146.16%
10eBr60.086 ± 0.0290.45%
10fMe16.16 ± 0.8328.83%
10gMe22.07 ± 0.2931.25%
10hMe31.78 ± 0.4733.66%
10iMe40.59 ± 0.1135.49%
10jMe50.38 ± 0.0540.23%
10kMe60.16 ± 0.0370.69%
Epacadostat--0.064 ± 0.01-
SAHA---91.66%
5a--6.52 ± 0.45-
a IC50 values are the means ± S.D. of three independent experiments.
Table 2. Inhibition activity of selected compounds against HDAC subtypes.
Table 2. Inhibition activity of selected compounds against HDAC subtypes.
IC50 (nM) a
Compd.HDAC1HDAC4HDAC6HDAC11SI c
7a244931.53% b84.446064.0029.00
10e10789.58%58.2324.37%18.51
10k11889.68%390.7029.77%3.04
SAHA90.8733,91137.1752,903.002.44
a IC50 performed using 10-dose IC50 mode with 3-fold serial dilution beginning from 10 μM. b % inhibition rate at 10 μM. c Selectivity Index (SI) = HDAC1 IC50 value/HDAC6 IC50 value.
Table 3. In vitro antiproliferative activity of selected compounds.
Table 3. In vitro antiproliferative activity of selected compounds.
IC50 (μM) a
Compd.HCT-116SW480MDA-MB-231MCF-7HK2
7a>80>80>80>80-
10d24.43 ± 4.78>80>8052.94 ± 6.95-
10e16.42 ± 3.7623.43 ± 3.8643.74 ± 1.8936.46 ± 5.45>80
10j>80>80>80>80-
10k>80>80>80>80-
SAHA2.64 ± 4.264.32 ± 2.2618.32 ± 5.616.32 ± 0.8826.60 ± 4.31
Epacadostat>80>80>80>80-
5a>80>80>80>80>80
a IC50 values are the means ± S.D. of three separate experiments.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, S.; Wang, Y.-F.; Lu, H.-R.; Yang, X.-Q.; Zhang, Y.; Ma, X.-L.; Huang, R.-Z. Discovery of Novel Imidazothiazole-Based Hydroxamic Acid Derivatives as Potent Indoleamine 2,3-Dioxygenase 1 and Histone Deacetylase 6 Dual Inhibitors. Molecules 2025, 30, 2508. https://doi.org/10.3390/molecules30122508

AMA Style

Zhang S, Wang Y-F, Lu H-R, Yang X-Q, Zhang Y, Ma X-L, Huang R-Z. Discovery of Novel Imidazothiazole-Based Hydroxamic Acid Derivatives as Potent Indoleamine 2,3-Dioxygenase 1 and Histone Deacetylase 6 Dual Inhibitors. Molecules. 2025; 30(12):2508. https://doi.org/10.3390/molecules30122508

Chicago/Turabian Style

Zhang, Shi, Yan-Fei Wang, Hai-Rui Lu, Xue-Qin Yang, Ye Zhang, Xian-Li Ma, and Ri-Zhen Huang. 2025. "Discovery of Novel Imidazothiazole-Based Hydroxamic Acid Derivatives as Potent Indoleamine 2,3-Dioxygenase 1 and Histone Deacetylase 6 Dual Inhibitors" Molecules 30, no. 12: 2508. https://doi.org/10.3390/molecules30122508

APA Style

Zhang, S., Wang, Y.-F., Lu, H.-R., Yang, X.-Q., Zhang, Y., Ma, X.-L., & Huang, R.-Z. (2025). Discovery of Novel Imidazothiazole-Based Hydroxamic Acid Derivatives as Potent Indoleamine 2,3-Dioxygenase 1 and Histone Deacetylase 6 Dual Inhibitors. Molecules, 30(12), 2508. https://doi.org/10.3390/molecules30122508

Article Metrics

Back to TopTop