Skip to main content
Journal of Enzyme Inhibition and Medicinal Chemistry logoLink to Journal of Enzyme Inhibition and Medicinal Chemistry
. 2025 Nov 10;40(1):2585606. doi: 10.1080/14756366.2025.2585606

Structure-based identification of a non-covalent thioredoxin reductase inhibitor with proven ADMET suitability

Giuseppe Lamanna a,b,#, Giuseppa Augello c,#, Luisa Ronga d, Diego Tesauro e,f, Ilaria Silvestri g, Antonina Azzolina c, Melchiorre Cervello c, Giuseppe Felice Mangiatordi b,, Michele Saviano g,
PMCID: PMC12604116  PMID: 41211658

Abstract

Thioredoxin reductase 1 (TrxR1), a selenoprotein enzyme crucial for redox homeostasis in mammals, has emerged as a promising target for anticancer therapy. In this study, we present a non-covalent TrxR1 inhibitor, identified through an integrated experimental and computational approach. After identifying a plausible druggable cavity, a molecular docking-based virtual screening of over 90,000 lead-like compounds was performed. The selected compounds were evaluated for their impact on TrxR1 activity, leading to the identification of the most promising candidate, C55. The identified compound, already proven to be free from potential ADMET concerns, inhibits TrxR1 in a dose-dependent manner, with an IC50 value in the micromolar range. C55′s activity was confirmed across multiple cancer cell lines, including HepG2, Huh7, MCF-7, and MDA-MB-231 cells. As a metal-free organic molecule capable of non-covalently inhibiting TrxR1, C55 represents a significant breakthrough, offering a solid foundation for hit-to-lead optimisation and the development of new anticancer drug candidates.

Keywords: Thioredoxin reductase 1, virtual screening, non-covalent inhibitor, anticancer therapy

GRAPHICAL ABSTRACT

graphic file with name IENZ_A_2585606_UF0001_C.jpg

Introduction

Thioredoxin reductases (TrxRs) are crucial enzymes in maintaining cellular redox balance, playing a key role in protecting against oxidative stress and regulating protein function through the thioredoxin system1. TrxRs are flavoproteins containing the 21st encoded amino acid selenocysteine2 (Sec, U), which is essential for the protein’s reducing activity. Three TrxRs are present in mammals: thioredoxin reductase 1 (TrxR1), thioredoxin reductase 2 (TrxR2), and thioredoxin reductase 3 (TrxR3). In numerous tumour types, the overexpression of cytosolic TrxR1 highlights its pivotal role in cancer cell survival, particularly in breast cancer (BCa)34, and hepatocellular carcinoma (HCC) cells5. Unlike normal cells, cancer cells exhibit heightened dependence on TrxR1 activity due to the increased oxidative stress induced by their altered metabolic processes. Targeting TrxR1 could disrupt their ability to manage this stress, presenting a compelling strategy for therapeutic intervention. Elevated expression of TrxR1 has been associated with a poor prognosis in patients with BCa and HCC as well as an increased risk of recurrence4, indicating that the enzyme may play a role in tumour progression and treatment resistance6. Thus, TrxR1 is a promising therapeutic target, as its inhibition could reduce tumour proliferation and increase sensitivity to conventional therapies7.

Recent studies have therefore focused on the design of selective TrxR1 inhibitors, which could limit the ability of tumour cells to maintain their favourable redox environment, thereby increasing their vulnerability to treatment and improving outcomes in patients with tumours with high TrxR1 expression7–10. Such therapeutic approaches could represent a new frontier in treating tumours resistant to conventional drugs. For all these reasons, the inhibition of TrxR1 has emerged as a promising strategy for cancer treatment, in recent years11–14. In particular, the most significant and well-studied class of TrxR1 inhibitors is represented by gold-containing compounds12,15–19. Initially recognised within the medicinal chemistry community for their efficacy in treating rheumatoid arthritis (RA) and tuberculosis20,21, these gold-based compounds have recently been explored as anticancer agents due to their cytotoxic activity across various cancer cell types22,23. One noteworthy example is represented by Auranofin, a gold(I)-containing compound initially approved as an antiarthritic drug, which has since been repurposed for cancer treatment22,24. In ovarian cancer, Auranofin demonstrates cytotoxic effects linked to its ability to inhibit the antioxidant activity of TrxR1. This inhibition reduces the removal rate of peroxides, leading to the accumulation of mitochondrial H2O2 and subsequent cell damage25,26. As an alternative to gold, platinum-based compounds have been employed for TrxR1 inhibition. For instance, cisplatin [cis-diamminedichloridoplatinum (II)] is a well-known platinum-based compound that has been approved for cancer treatment27,28. Notably, cisplatin, in addition to acting on the bases of DNA29 has demonstrated highly specific and irreversible inhibition of TrxR130. Another compound known to inhibit TrxR1 is arsenic trioxide31, which has been established as an effective treatment for acute promyelocytic leukaemia32. More recently, a rhodium(I) N-heterocyclic carbene complex has also been described to inhibit TrxR, increase intracellular ROS accumulation, and promote apoptosis in HCC cells5. In general, these metal-based complexes are designed to inhibit TrxR1 through the covalent modification of the catalytic Sec, displaying a high affinity towards metals33. In a similar way, β-unsaturated carbonyl compounds known as Michael acceptors could inhibit TrxR1 by covalent interaction with the Sec34. Although these molecules are undoubtedly interesting for developing new therapeutic strategies based on TrxR1 inhibition, they have several significant limitations. First, most of them contain metals, raising concerns about potential toxicity. Gold-based compounds, for instance, can cause nephrotoxicity, allergic dermatitis, and, in some cases, myelotoxicity35. Platinum-based compounds, although still in use, are known to cause significant renal damage, peripheral neuropathy, hearing loss, and myelotoxicity (bone marrow suppression)36,37. Noteworthy, most of the compounds identified so far as TrxR1 inhibitors covalently modify the protein. Identifying non-covalent inhibitors of TrxR1 could offer several advantages in terms of: i) specificity: covalent inhibitors of TrxR1 target selenocysteine sharing a similar reactivity with cysteine which is a more common amino acid in proteins, potentially leading to off-target effects; ii) pharmacokinetics properties: non-covalent inhibitors provide a more tuneable duration of action; iii) easier optimisation: non-covalent compounds can be more easily optimised through structural modifications to refine their affinity, selectivity, and bioavailability without relying on covalent interaction with a specific residue.

The present work represents a first attempt to address the limitations of the covalent inhibition of TrxR1 through an integrated computational/experimental approach. Using a workflow combining cavity mapping, molecular docking, direct binding assays, and cell-based assays, we have developed a virtual screening (VS) protocol for identifying compounds capable of non-covalently inhibiting TrxR1. Remarkably, to ensure that the selected compounds are free from potential Adsorption Distribution Metabolism Excretion and Toxicity (ADMET) issues and, therefore, suitable for rapid hit-to-lead optimisation, the screening was conducted on the Asinex Elite library, which comprises more than 90,000 compounds that have already been pre-screened against a panel of early ADMET tests. The study enabled us to identify a new compound with notable pharmacokinetic properties that inhibits TrxR1 non-covalently. The paper is discussed in the perspective of using this compound as starting point for subsequent rational optimisation studies. Notice that this study has previously made available as a preprint38.

Materials and methods

Cavity mapping and data retrieval

Cavity mapping was performed on the X-ray coordinates of recombinant TrxR1 with reduced C-terminal tail (PDB code: 3EAN)39. To this aim, we used the SiteMap40 tool, available in the Schrodinger suite 2020–1. The protein was prepared using the protein preparation wizard tool available in the Schrödinger Suite41,42. This preparation involved determining protonation states at pH 7.4, minimising energy, and removing water molecules. All the ligands were prepared using the LigPrep tool43 employing the OPLS5 force field for energy minimisation44, and a maximum of 32 stereoisomers for each ligand. Tautomeric forms at pH 7.4 were also generated.

Docking simulations

Docking simulations were performed using the GOLD docking software45. GOLD employs a genetic algorithm for conformational sampling and defines its own performance metrics. All genetic algorithm parameters were set to auto so that GOLD was free to find the optimal parameters for the specific calculation. Building on the performed cavity mapping, the box was centred on the atom number 376 (radius = 10 Å), which is the selenium atom in the selenocysteine 498 residue. GOLD was instructed to restrict the binding site definition to concave, solvent-accessible surfaces using the LIGSITE algorithm46. Chemscore47 was used as a scoring function. Note that, in addition to the Chemscore value, its binding energy term was also reported, as recommended in the original paper by Eldridge et al.48

Virtual screening

A docking-bases VS campaign was conducted on the entire Asinex Elite dataset49, which includes 91,001 commercially available ‘lead-like’ compounds. These compounds underwent experimental screening against an initial set of ADMET tests to confirm they were free of potential ADMET issues and suitable for rapid hit-to-lead optimisation. Each compound was prepared and docked into the crystal structure, as described. The compounds were then divided into ten groups, and the docking was run in parallel on an HP Z4 workstation, a strategy that significantly optimised computational time. The compounds were then ranked based on the returned ChemScore, and the top 500 poses were selected for the next steps. The binding modes of the top 500 compounds underwent rigorous visual inspection, a crucial step in the decision-making process following a virtual screening (VS) procedure50. 63 compounds were finally selected for further experimental testing.

Cell lines and cell culture

Human HCC HepG2 (HB-8065TM) cells were obtained from the American Type Culture Collection (ATCC, Rockville, MD). Cells were maintained in Roswell Park Memorial Institute (RPMI) 1640 medium (MERCK, Milan, Italy) containing 10% (v/v) Foetal Bovine Serum (FBS) (Gibco-Thermo Fisher Scientific, Milano, Italy), 1 mM sodium pyruvate (MERCK), 100 units/ml penicillin (MERCK) and 100 µg/ml streptomycin (MERCK). The HCC Huh7 cell line was a gift from Professor Massimo Levrero (Sapienza University of Rome, Italy) and was maintained in a high-glucose Dulbecco’s Modified Eagle Medium (DMEM - MERCK) containing 10% FBS. The human breast cancer cell lines MCF-7 and MDA-MB-231 were obtained from ATCC (HTB-22 and HTB-26, respectively) and were maintained in high-glucose DMEM medium containing 10% FBS and antibiotics.

Cell viability assay

Cells (5 x 103/well) were seeded in 96-well plates and, after 24 h, were exposed to the selected Asinex Elite compounds at different concentrations from 1 to 100 µM. It is worth noting that in the case of chiral compounds, a mixture of stereoisomers was tested since the supplier was not able to provide enantiomerically pure compounds. After 72 or 96 h, MTS assays were performed using the CellTiter Aqueous OneSolution kit (Promega Corporation Madison, WI, USA) according to the manufacturer’s instructions. Cell viability was expressed as a percentage of the absorbance measured in the control cells. Values were expressed as means ± Standard Deviation (SD) of at least two experiments, each performed in triplicate.

Measurement of TrxR1 activity

TrxR1 activity was assessed using the Thioredoxin Reductase Assay Kit according to the manufacturer’s instructions (Sigma-Aldrich, Milan, Italy). The TrxR1 enzyme, extracted from rat liver, was supplied with the kit (catalog number CS0170), and was provided in ready-to-use form by the manufacturer. In brief, various concentrations of SYN19823426 (C55) compound and Auranofin (Sigma-Aldrich) were incubated with TrxR1 extracted from rat liver and added to each well of 96-well microtiter plates in the presence of NAPDH and 5,5′-dithio-bis(2-nitrobenzoic acid) (DTNB) at 25 °C for 60 min. TrxR catalyses the reduction of DTNB with NADPH to 5-thio-2-nitrobenzoic acid (TNB), which produces an intense yellow colour. Therefore, TrxR1 activity was spectrophotometrically determined at 412 nm absorbance using a microplate reader (Bio-Rad Laboratories, CA, USA). Values were expressed as a percentage of activity compared to solvent alone ± SD of at least two separate experiments. Furthermore, the determination of TrxR1 activity was performed on HepG2, Huh7, MCF-7 and MDA-MB-231 whole cell lysates. Briefly, exponentially growing adherent cells were lysed by adding ice-cold Radio-Immunoprecipitation Assay (RIPA) buffer (Cell Signalling Technologies Inc., Beverly, MA, USA) containing proteases and phosphatase inhibitors (Sigma-Aldrich) for 20 min on ice. Lysates were centrifuged, and protein concentration in supernatants was quantified by a Protein assay kit (Bio-Rad). Therefore, 10 μg of proteins were used to determine TrxR1 activity in cell lysates using the Thioredoxin Reductase Assay Kit as reported above. The values were expressed as a percentage of activity compared to solvent alone ± SD of at least two separate experiments using two whole cell lysates for each cell line.

Measurement of Trx1 activity

Thioredoxin-1 (Trx1) activity was measured using the ProteoStat Thioredoxin-1 assay kit (Enzo Life Sciences, Plymouth Meeting, PA, USA) according to the manufacturer’s protocol. Briefly, 10 μL of the test compound C55, at different concentrations, was added to each well of a black 96-well microplate. For control, 10 μL of Aurothiomalate working solution (provided in the kit and prepared as recommended) was dispensed into designated wells. Subsequently, 70 μL of Thioredoxin-1 Assay Master Mix and 10 μL of dithiothreitol (DTT) were added to each well. The plate was incubated for 30 min at room temperature in the dark. The reaction was stopped by adding 10 μL of the Stop Reagent working solution to each well, followed by gentle mixing for ∼30 s. Fluorescence was measured using a GloMax® microplate reader (Promega) at an excitation wavelength of 500 nm and an emission wavelength of 603 nm.

RNA extraction and real-time PCR

Total RNA was extracted using the TRIzol method and 1 μg of RNA was used for reverse transcription to generate cDNA using the QuantiNova Reverse Transcription Kit (QIAGEN), following the instructions of the manufacturer. Quantitative Real-time PCR was analysed using the QuantiNova SYBR Green PCR Kit (QIAGEN) and QuantiTect primers (QIAGEN) specific for NQO1 (QT00050281), HO-1 (QT00092645) and β-actin (QT00095431). Data are expressed as the relative mRNA expression level of the different genes in treated cells compared with control cells. Values given are the mean ± SD of three different experiments performed in triplicate.

Intracellular ROS detection

Cells were seeded in 96-well plates and after 24 h were exposed to C55 or Auranofin. The cells were washed with phosphate-buffered saline (PBS) prior to probing with 40 μM H2DCFDA (Invitrogen, Milan, Italy) for 1 h at 37 °C with 5% CO2 in the dark. Excess H2DCFDA was removed with PBS. The fluorescence intensity was measured using a GloMax® microplate reader, with excitation and emission wavelengths of 485 nm and 530 nm, respectively. The fluorescence intensity of dichlorofluorescein (DCF) was calculated as the fluorescence intensity of treated cells relative to non-treated cells.

Western blotting

RIPA buffer (Cell Signalling Technologies Inc.) was used to obtain cell lysates, and Western blotting was performed using the methodology for the Odyssey® infra-red imaging system (LI-COR Biosciences, NE, USA). After transfer nitrocellulose membranes were placed in Odyssey® blocking buffer (OBB, LI-COR) diluted in tris-buffered saline (TBS) and incubated for 1 h at room temperature. Primary antibodies were diluted in OBB. Secondary antibodies conjugated to IRDye® 800CW (LI-COR) or Alexa Fluor 680 (Molecular Probes, Invitrogen Carlsbad, CA, USA) were diluted in OBB. Membranes were scanned and analysed with an Odyssey IR scanner using Odyssey 3.0 imaging software. The relative expressions were calculated as the ratio of drug-treated samples vs. control (DMSO) and corrected using the quantified level of β-actin expression. Primary antibodies raised against γ-H2AX and β-actin were purchased from Cell Signalling Technologies and Sigma-Aldrich, respectively.

C55 stability and integrity assessment

The identity and stability of compound C55 in DMSO over one month were evaluated by Electrospray ionisation (ESI) mass spectrometry. Samples were prepared by diluting 1 µL of a 10 mM C55 solution in DMSO with 100 µL of methanol. Spectra were recorded in positive‐ion mode on an Applied Biosystems mass spectrometer equipped with a triple‐quadrupole analyser. The observed molecular‐ion peak confirmed both the integrity and the unchanged mass of C55 throughout the ageing period (see Figure S1 in the Supplementary Information).

Results and discussion

Pocket recognition

As a first step of the study, we searched for a binding site on the TrxR1 surface that could accommodate small molecules (SMs). The information regarding the protein portion responsible for the SMs accommodation is, in fact, still elusive with the few structure-based studies present in the literature that are not aligned regarding its exact localisation51,52. The X-ray 3D structure of the protein published by Cheng et al. (PDB code: 3EAN39) was used as input for Sitemap. Among the five detected cavities, P1, shown in Figure 1, was predicted to be the most promising one, as it returned the highest SiteScore (1.039) and DScore (1.075) values. The identified cavity, nestled between chains A and B of the protein and in proximity to the residue selenocysteine in position 498 (U498), holds significant promise. The computed SiteScore and DScore values, characteristic of sites with high druggability potential40,47, underscore the potential impact of our findings on structure-based drug design approaches.

Figure 1.

Figure 1.

Druggable cavity (P1) identified by SiteMap on the dimer of the TrxR1 protein structure (PDB code: 3EAN)39. The two monomers are designated as chain A (green) and chain B (blue), while P1 is shown as a grey surface. The selenocysteine U498 of chain A is highlighted using a stick representation.

Virtual screening

To identify SMs able to target the chosen cavity, we performed a VS of a large chemical library using the software GOLD45, available as a Cambridge Crystallographic Data Centre (CCDC) product. All the compounds were docked on the identified TrxR1 binding site (P1) employing as protein structure the one published by Cheng et al.39 Based on the returned ChemScore, the top 500 compounds were selected and the corresponding top-scored poses visually inspected. We finally selected 63 initial hits for further in-vitro experimental tests (Figure 2).

Figure 2.

Figure 2.

2D structures and Asinex title of the 63 compounds selected based on the performed VS procedure. The molecule selected as a TrxR inhibitor is highlighted with a red box, while the other molecules that showed interesting cytotoxicity values are highlighted with a black box.

Cell-based screening of asinex library compounds

Only 37 of the 63 compounds selected from the Asinex chemical library were completely soluble in DMSO at a concentration of 50 mM and therefore tested for their effect on cell cytotoxicity against human HCC and BCa cell lines. Initial screening was conducted by MTS assay for 72 h at concentrations of 10, 50 and 100 µM, and only samples found to have cytotoxic effects at a concentration ≤ 50 µM on at least 2 out of 4 cell lines were used for further analysis (Table 1).

Table 1.

IC50 values of the 37 compounds against HepG2, Huh7, MCF-7 and MDA-MB-231 cells for 72h of treatment.

    IC50 (µM)
compound number compound name HepG2 Huh7 MCF-7 MDA-MB-231
C4 SYN22041065 96.3 ± 6.6 60.5 ± 10.2 85.5 ± 7.2 > 100
C5 SYN19990927 > 100 > 100 > 100 > 100
C18 AEM10027871 > 100 > 100 > 100 > 100
C21 LEG19990218 28.5 ± 9.8 25.7 ± 1.8 70.1 ± 3.0 51.4 ± 7.8
C22 LEG20009354 > 100 > 100 > 100 > 100
C23 LEG15341540 60.5 ± 1.8 51.7 ± 15.41 > 100 > 100
C24 LMK17221003 48.6 ± 15.3 50.2 ± 25.5 > 100 > 100
C25 LEG18687308 > 100 > 100 > 100 > 100
C26 SYN17860685 > 100 > 100 > 100 > 100
C27 ASF192797 50 21.8 ± 2.3 51.2 ± 12.5 > 100 > 100
C28 ASF19632166 > 100 > 100 > 100 > 100
C29 ADM19835767 > 100 > 100 > 100 > 100
C30 AOP18687354 > 100 > 100 > 100 > 100
C31 SYN15659685 > 100 > 100 > 100 > 100
C33 SYN15643124 > 100 > 100 > 100 > 100
C34 LEG17851037 > 100 > 100 > 100 > 100
C35 SYN17920976 > 100 > 100 > 100 > 100
C36 SYN17736818 > 100 > 100 > 100 > 100
C37 LMG18101294 > 100 > 100 > 100 > 100
C39 AEM16401255 63.4 ± 12.3 23.9 ± 12.1 81.0 ± 9.4 > 100
C40 ASF17521182 61.7 ± 24.5 78.55 ± 19.1 > 100 > 100
C41 AEM18663974 18.8 ± 14.7 > 100 48.9 ± 15.7 55.13 ± 11.1
C42 AEM18511305 45.9 ± 1.8 56.4 ± 12.5 49.2 ± 16.6 > 100
C44 ASF19865209 70.4 ± 25.5 72.9 ± 18.5 > 100 > 100
C45 LEG19971843 > 100 98.2 ± 11.1 > 100 > 100
C47 SYN13244281 54.1 ± 5.4 27.7 ± 9.0 49 ± 1.1 > 100
C49 SYN22976891 > 100 > 100 > 100 > 100
C50 LEG15394831 > 100 60.2 ± 11.0 > 100 > 100
C51 ASF20692226 > 100 73.1 ± 21.0 72.1 ± 12.5 > 100
C52 AOP20989282 > 100 > 100 > 100 > 100
C53 AOP22041441 61.1 ± 18.2 72.6 ± 20.0 > 100 > 100
C54 SYN22964503 > 100 > 100 > 100 > 100
C55 SYN19823426 29.4 ± 5.3 29.9 ± 3.6 > 100 42.7 ± 1.6
C57 LEG19412213 > 100 > 100 > 100 > 100
C58 IPE21905364 61.4 ± 12.2 24.4 ± 12.0 43.2 ± 22.0 > 100
C59 SYN20054602 > 100 > 100 > 100 > 100
C60 ASF18586547 > 100 > 100 > 100 > 100

Cell-free and cell lysates thioredoxin reductase 1 activity

Following the cell-based screening, we identified six compounds (C21, C41, C42, C47, C55, and C58) for further investigation. Our initial tests in a cell-free assay, with all compounds tested at concentrations up to 10 µM, revealed that five of the six compounds showed very low (less than 25%) or no inhibitory activity against TrxR1. However, compound C55 (SYN19823426) (Figure 3) stood out.

Figure 3.

Figure 3.

Inhibition of TrxR1 activity by selected compounds in cell-free assay. Rat liver TrxR1 and were incubated with 10 µM of each compound and 0.5 µM of Auranofin (AUR). TrxR1 activity was assayed as described in material and methods. Values are expressed as percentage of activity compared to solvent alone (-).

We then proceeded to test the inhibitory activity of compound C55 on TrxR1 in a dose-response experiment. As shown in Figure 4(A), C55 inhibited TrxR1 activity in a dose-dependent manner, with an IC50 of 4.7 µM ± 1.3. Furthermore, we extended our evaluation to protein samples extracted from HepG2, Huh7 MCF-7, and MDA-MB-231 cells. Figure 4(B) illustrates that C55′s inhibitory activity on TrxR1 varied depending on the cell type, underscoring the need for further investigation into its potential differential effects. To ensure that the measured enzymatic activity reflected TrxR1-specific activity and was not influenced by low-molecular-weight thiols, we performed the assay in the presence of Auranofin, a selective TrxR1 inhibitor. As shown in Figure S2, the marked reduction in activity upon Auranofin treatment confirmed the specificity of the assay for TrxR1. Furthermore, to investigate the specificity of the interaction between C55 and TrxR1, particularly with respect to potential interference from small molecule thiols, we conducted a thioredoxin 1 (Trx1) activity assay in the presence of the compound, since this protein is known to contains thiol groups in its active site. Aurothiomalate, a known Trx1 inhibitor, was included as a positive control. The results obtained showed that C55 did not inhibit Trx1 activity (Figure S3). These data support the idea that C55 specifically acts on the TrxR1 reductase and probably on its C-terminal Sec-containing motif.

Figure 4.

Figure 4.

Inhibition of TrxR1 activity by C55 in vitro. A) rat liver TrxR1 and B) 10 µg of protein samples extracted from human HCC (HepG2, Huh7) or BCa (MCF-7, MDA-MB-231) cells were incubated with the indicated concentrations of C55 and then TrxR1 activity was assayed as described in material and methods. Kit inhibitor was used as positive control inhibitor for TrxR1. Values are expressed as percentage of activity compared to solvent alone ± SD of at least two separate experiments.

Effect of C55 on cell viability of HCC and BCa cell lines

Next, we examined the effect of C55 on the cell viability of HCC (HepG2, Huh7) and BCa (MCF-7, MDA-MB-231) cell lines by MTS assay. As shown in Figure 5, C55 reduced cell viability in a dose-and time-dependent manner in all cell lines. To further support the involvement of the thioredoxin system in cell viability, we evaluated the effect of Auranofin, a known TrxR1 inhibitor, on the same cell lines. As shown in Figure S4, Auranofin treatment led to a marked reduction in cell viability, consistent with the proposed role of TrxR1 inhibition in mediating the cytotoxic effects observed with C55. To assess the reversibility of TrxR1 inhibition, a washout viability assay was performed: HepG2 and MDA-MB-231 cells were incubated with the inhibitor C55 for 24 h, followed by medium replacement with fresh compound-free medium and further incubated for 48 h (Figure S5). A recovery in cell viability was observed after the washout, indicating that the inhibitory effect of C55 is reversible. In contrast to C55 treatment, Auranofin-treated cells did not recover viability upon washout, consistent with its irreversible mode of action. These results are shown in Figure S5.

Figure 5.

Figure 5.

Effects of C55 treatment on cell viability in HCC (HepG2, Huh7) and BCa (MCF-7, MDA-MB-231) cell lines. Cells were treated with different concentrations of C55 for 72 (A) or 96 (B) hours. Cell viability was determined by using MTS assay.

However, different cell lines show different sensitivity to the compound, with MCF-7 cells showing the lowest sensitivity, as shown by the IC50 values in Table 2.

Table 2.

IC50 values of compound C55 against HepG2, Huh7, MCF-7, and MDA-MB-231 cells after 72 and 96h of treatment.

hours IC50 (µM)
HepG2 Huh7 MCF-7 MDA-MB-231
72 29.4 ± 5.3 29.9 ± 3.6 > 100 42.7 ± 1.6
96 9.3 ± 3.7 18.9 ± 8.7 > 100 29.5 ± 10.6

Effects of C55 on oxidative stress signalling

Inhibition of TrxR1 increases intracellular oxidative stress, triggering activation of the of antioxidant genes, such as NAD(P)H:quinone oxidoreductase 1 (NQO1) and haem oxygenase-1 (HO-1)53,54. As shown in Figure 6(A), treatment with C55 for 48 h significantly upregulated the expression levels of these two genes in HepG2 and MDA-MB-231 cells, as analysed by real-time PCR.

Figure 6.

Figure 6.

Effects of C55 on oxidative stress signalling. A) Gene expression analysis by real-time PCR in HepG2 and MDA-MB-231 cells treated for 48 h with the indicated concentrations of C55 and Auranofin (AUR). *p < 0.05, **p < 0.01, C55 or AUR versus control vehicle alone B) ROS generation in HepG2 and MDA-MB-231 cells treated with C55. Cells were untreated or treated with C55 or AUR at the indicated concentrations for 48 h, and intracellular ROS levels were evaluated using H2DCFDA as a probe. Data are expressed as the percentage of control cells and are the means ± SD of two separate experiments, each performed in triplicate. * p < 0.05, C55 or AUR versus control vehicle alone. C) Western blotting analysis of γ-H2AX in HepG2 and MDA-MB-231 cells treated for 48 h with the indicated concentrations of C55 and AUR.

C55 treatments also increased intracellular reactive oxygen species (ROS) production, as measured by using the cell-permeable fluorescent probe H2DCFDA (Figure 6B). Furthermore, since high levels of ROS are known to produce DNA damage, we next evaluated the effect of C55 on the expression levels of phospho-H2AX (γ-H2AX) histone, a marker of DNA damage55 (Figure 6(C)). Treatments of HepG2 and MDA-MB-231 cells with C55 for 48 h increased γ-H2AX expression levels, indicating genotoxic stress. Auranofin produced similar effects (Figures 6(B,C)). These findings support that C55 acts as a TrxR1 inhibitor, promoting oxidative stress and DNA damage in cancer cells.

A new binding mode for TrxR1 inhibition

The experimental data, taken together, put forward compound C55 as a SM acting as non-covalent TrxR inhibitor. It is worth noting, in fact, that such a compound lacks groups that can covalently interact with selenocysteine, is metal-free, and is characterised by a novel chemotype.

Furthermore, the binding mode returned by GOLD (Figure 7), does not involve covalent interactions. Nonetheless, the (S,S) configuration of C55 shows a promising docking score (ChemScore of 36.16 KJ/mol) supported by a favourable estimated binding energy of −37.55 KJ/mol.

Figure 7.

Figure 7.

Top-scored docking pose of C55 within the TrxR1 binding site (PDB code: 3EAN)39.

The hypothesis returned by the pocket prediction phase is here confirmed, as the experimentally validated molecule is predicted to interact with the hypothesised residues, namely G499 from chain A and Y116 from chain B, with the addition of the selenium-containing residue U498, validating all the previously proposed hypotheses and in agreement with previous knowledge56. In summary, the following clues can be derived based on the discussed data: (i) the pocket prediction algorithm correctly predicted the location of the cavity to be considered; (ii) the selenium-containing residue plays a relevant role in the binding mode, establishing a hydrogen bond involving its backbone and confirming its already known role in the inhibition of the enzyme57; (iii) the virtual screening procedure identified 63 compounds, among which compound C55 showed an inhibitory activity with an IC50 of 4.7 ± 1.3 µM; (iv) the proposed compound is devoid of pharmacokinetic hindrances by hypothesis, as it belongs to a commercial library comprising only compounds devoid of potential ADMET problems49. This compound, to the best of our knowledge, is the first metal-free compound able to bind to TrxR with a non-covalent interaction, making it a significant starting point for further studies aimed at its optimisation and, hopefully, at the identification of unexplored molecular series capable of efficiently inhibiting such a promising anticancer target. Noteworthy, the identified initial hit has never been studied as a bioactive molecule, as evident after interrogating ChEMBL58,59 and PubChem60 repositories. This result is even more significant considering that the compound has already undergone experimental ADMET tests, which confirmed the absence of pharmacokinetic issues.

Conclusions

This study led to the identification of an interesting non-covalent inhibitor of Thioredoxin reductase 1 (TrxR1), a selenoprotein enzyme crucial for maintaining redox homeostasis in mammals. Through an integrated experimental and computational approach, the compound C55 was identified as a promising candidate, demonstrating dose-dependent inhibition of TrxR1 with an IC50 value in the micromolar range. The identified compound, already proved to be free of ADMET-related issues, exhibited inhibitory activity across various cancer cell lines, including HepG2, Huh7, MCF-7, and MDA-MB-231. As the first metal-free organic molecule to act as a non-covalent inhibitor of TrxR1, C55 represents a significant step forward, providing a strong foundation for hit-to-lead optimisation and paving the way for the development of potent anticancer drug candidates.

Supplementary Material

Supplementary_information_ Clean.docx

Acknowledgements

Luisa Ronga acknowledges “La ligue contre le cancer” for supporting this work. The graphical abstract was created using BioRender.com. GL wrote the original draft of the manuscript and produced/analysed the computational data; GA, AA and CM wrote the original draft of the manuscript and produced/analysed the experimental data; LR, DT and IS analysed data, reviewed and edited the manuscript; GFM analysed data, wrote the original draft of the manuscript and supervised the research; MS reviewed and edited the manuscript, supervised the research. All authors had edited and approved the final manuscript.

Funding Statement

This work was supported grant by the Italian National Research Council (CNR) “The Bioinorganic Drugs (BIDs) joint laboratory: A multidisciplinary platform promoting new molecular targets for drug discovery” is acknowledged, and by the research project “Potentiating the Italian Capacity for Structural Biology Services in Instruct Eric” (Acronym: ITACA.SB, project n° IR0000009) within the call MUR D.D. 0003264 dated 28/12/2021 PNRR M4/C2/L3.1.1, funded by the European Union NextGenerationEU.

Author contributions statement

CRediT: Giuseppe Lamanna: Data curation, Formal analysis, Methodology, Writing – original draft; Giuseppa Augello: Data curation, Formal analysis, Writing – original draft; Luisa Ronga: Formal analysis, Writing – review & editing; Diego Tesauro: Formal analysis, Writing – review & editing; Ilaria Silvestri: Formal analysis, Writing – review & editing; Antonina Azzolina: Formal analysis, Methodology, Writing – original draft; Cervello Melchiorre: Formal analysis, Investigation, Methodology, Writing – original draft; Giuseppe Felice Mangiatordi: Conceptualization, Data curation, Formal analysis, Methodology, Supervision, Writing – original draft, Writing – review & editing; Michele Saviano: Funding acquisition, Supervision, Writing – review & editing.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author/s.

References

  • 1.Pennington JD, Jacobs KM, Sun L, Bar-Sela G, Mishra M, Gius D.. Thioredoxin and thioredoxin reductase as redox-sensitive molecular targets for cancer therapy. Curr Pharm Des. 2007;13(33):3368–3377. [PubMed] [Google Scholar]
  • 2.Tamura T, Stadtman TC.. A New selenoprotein from human lung adenocarcinoma cells: purification, properties, and thioredoxin reductase activity. Proc Natl Acad Sci U S A. 1996;93(3):1006–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Patwardhan RS, Rai A, Sharma D, Sandur SK, Patwardhan S.. Txnrd1 as a prognosticator for recurrence, metastasis and response to neoadjuvant chemotherapy and radiotherapy in breast cancer patients. Heliyon. 2024;10(6):e27011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bhatia M, McGrath KL, Di Trapani G, Charoentong P, Shah F, King MM, Clarke FM, Tonissen KF.. The thioredoxin system in breast cancer cell invasion and migration. Redox Biol. 2016;8:68–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fan R, Bian M, Hu L, Liu W.. A new rhodium(I) NHC complex inhibits TrxR: in vitro cytotoxicity and in vivo hepatocellular carcinoma suppression. Eur J Med Chem. 2019;183:111721. [DOI] [PubMed] [Google Scholar]
  • 6.López-Grueso MJ, González R, Muntané J, Bárcena JA, Padilla CA.. Thioredoxin downregulation enhances sorafenib effects in hepatocarcinoma cells. Antioxidants (Basel). 2019;8(10):501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gencheva R, Arnér ESJ.. Thioredoxin reductase inhibition for cancer therapy. Annu Rev Pharmacol Toxicol. 2022;62(1):177–196. [DOI] [PubMed] [Google Scholar]
  • 8.Lu Q, Shan M, Yang J, Xu Z, Lei Y, Liu W.. A Dinuclear gold(I) complex with Bis(N-Heterocyclic Carbene) ligands potentiated immune responses against liver cancer via ROS-driven endoplasmic reticulum stress and ferroptosis. Chin. Chem. Lett. 2025;36(12):110940. [Google Scholar]
  • 9.Lu Y, Liu Y, Liang Z, Ma X, Liu L, Wen Z, Tolbatov I, Marrone A, Liu W.. NHC-Gold(I)-alkyne complexes induced hepatocellular carcinoma cell death through bioorthogonal activation by palladium complex in living system. Chin. Chem. Lett. 2023;34(9):108413. [Google Scholar]
  • 10.Bian M, Fan R, Zhao S, Liu W.. Targeting the thioredoxin system as a strategy for cancer therapy: miniperspective. J Med Chem. 2019;62(16):7309–7321. [DOI] [PubMed] [Google Scholar]
  • 11.Ghareeb H, Metanis N.. The thioredoxin system: a promising target for cancer drug development. Chemistry. 2020;26(45):10175–10184. [DOI] [PubMed] [Google Scholar]
  • 12.Fereidoonnezhad M, Ahmadi Mirsadeghi H, Abedanzadeh S, Yazdani A, Alamdarlou A, Babaghasabha M, Almansaf Z, Faghih Z, McConnell Z, Shahsavari HR, et al. Synthesis and biological evaluation of thiolate gold (i) complexes as thioredoxin reductase (TrxR) and glutathione reductase (GR) inhibitors. New J Chem. 2019;43(33):13173–13182. [Google Scholar]
  • 13.Xu J, Arnér ESJ.. Pyrroloquinoline quinone modulates the kinetic parameters of the mammalian selenoprotein thioredoxin reductase 1 and is an inhibitor of glutathione reductase. Biochem Pharmacol. 2012;83(6):815–820. [DOI] [PubMed] [Google Scholar]
  • 14.Scalcon V, Bindoli A, Rigobello MP.. Significance of the mitochondrial thioredoxin reductase in cancer cells: an update on role, targets and inhibitors. Free Radic Biol Med. 2018;127:62–79. [DOI] [PubMed] [Google Scholar]
  • 15.Marzo T, Cirri D, Gabbiani C, Gamberi T, Magherini F, Pratesi A, Guerri A, Biver T, Binacchi F, Stefanini M, et al. Auranofin, Et3 PAuCl, and Et3 PAuI are highly cytotoxic on colorectal cancer cells: a chemical and biological study. ACS Med Chem Lett. 2017;8(10):997–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Quero J, Cabello S, Fuertes T, Mármol I, Laplaza R, Polo V, Gimeno MC, Rodriguez-Yoldi MJ, Cerrada E.. Proteasome versus thioredoxin reductase competition as possible biological targets in antitumor mixed thiolate-dithiocarbamate gold(III) complexes. Inorg Chem. 2018;57(17):10832–10845. [DOI] [PubMed] [Google Scholar]
  • 17.Reddy KM, Mugesh G.. Modelling the inhibition of selenoproteins by small molecules using cysteine and selenocysteine derivatives. Chemistry. 2019;25(37):8875–8883. chem [DOI] [PubMed] [Google Scholar]
  • 18.Tavares TT, Azevedo GC, Garcia A, Carpanez AG, Lewer PM, Paschoal D, Müller BL, Dos Santos HF, Matos RC, Silva H, et al. Gold(I) complexes with aryl-thiosemicarbazones: molecular modeling, synthesis, cytotoxicity and TrxR inhibition. Polyhedron. 2017;132:95–104. [Google Scholar]
  • 19.Bindoli A, Rigobello MP, Scutari G, Gabbiani C, Casini A, Messori L.. Thioredoxin reductase: a target for gold compounds acting as potential anticancer drugs. Coord. Chem. Rev. 2009;253(11-12):1692–1707. [Google Scholar]
  • 20.Shaw CF. Gold-based therapeutic agents. Chem Rev. 1999;99(9):2589–2600. [DOI] [PubMed] [Google Scholar]
  • 21.Kerimova AA, Atalay M, Yusifov EY, Kuprin SP, Kerimov TM.. Antioxidant enzymes; possible mechanism of gold compound treatment in rheumatoid arthritis. Pathophysiology. 2000;7(3):209–213. [DOI] [PubMed] [Google Scholar]
  • 22.Milacic V, Fregona D, Dou Q.. Gold complexes as prospective metal-based anticancer drugs. Histol. Histopathol. 2007;23(1):101–108. [DOI] [PubMed] [Google Scholar]
  • 23.Ronconi L, Giovagnini L, Marzano C, Bettìo F, Graziani R, Pilloni G, Fregona D.. Gold dithiocarbamate derivatives as potential antineoplastic agents: design, spectroscopic properties, and in vitro antitumor activity. Inorg Chem. 2005;44(6):1867–1881. [DOI] [PubMed] [Google Scholar]
  • 24.Roder C, Thomson MJ.. Auranofin: repurposing an old drug for a golden new age. Drugs R D. 2015;15(1):13–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Marzano C, Gandin V, Folda A, Scutari G, Bindoli A, Rigobello MP.. Inhibition of thioredoxin reductase by auranofin induces apoptosis in cisplatin-resistant human ovarian cancer cells. Free Radic Biol Med. 2007;42(6):872–881. [DOI] [PubMed] [Google Scholar]
  • 26.Rigobello MP, Folda A, Baldoin MC, Scutari G, Bindoli A.. Effect of auranofin on the mitochondrial generation of hydrogen peroxide. role of thioredoxin reductase. Free Radic Res. 2005;39(7):687–695. [DOI] [PubMed] [Google Scholar]
  • 27.Boulikas T, Vougiouka M.. Recent clinical trials using cisplatin, carboplatin and their combination chemotherapy drugs. Oncol Rep. 2004;11(3):559–595. [PubMed] [Google Scholar]
  • 28.Urig S, Becker K.. On the potential of thioredoxin reductase inhibitors for cancer therapy. Semin Cancer Biol. 2006;16(6):452–465. [DOI] [PubMed] [Google Scholar]
  • 29.Brabec V, Hrabina O, Kasparkova J.. Cytotoxic platinum coordination compounds. DNA binding agents. Coord. Chem. Rev. 2017;351:2–31. [Google Scholar]
  • 30.Arnér ESJ, Nakamura H, Sasada T, Yodoi J, Holmgren A, Spyrou G.. Analysis of the inhibition of mammalian thioredoxin, thioredoxin reductase, and glutaredoxin by Cis -diamminedichloroplatinum (II) and its major metabolite, the glutathione-platinum complex. Free Radic Biol Med. 2001;31(10):1170–1178. [DOI] [PubMed] [Google Scholar]
  • 31.Lu J, Chew E-H, Holmgren A.. Targeting thioredoxin reductase is a basis for cancer therapy by arsenic trioxide. Proc Natl Acad Sci U S A. 2007;104(30):12288–12293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mayorga J, Richardson-Hardin C, Dicke KA.. Arsenic trioxide as effective therapy for relapsed acute promyelocytic leukemia. Clin J Oncol Nurs. 2002;6(6):341–346. [DOI] [PubMed] [Google Scholar]
  • 33.Bernabeu De Maria M, Lamarche J, Ronga L, Messori L, Szpunar J, Lobinski R.. Selenol (-SeH) as a target for mercury and gold in biological systems: contributions of mass spectrometry and atomic spectroscopy. Coord. Chem. Rev. 2023;474:214836. [Google Scholar]
  • 34.Gan F-F, Kaminska KK, Yang H, Liew C-Y, Leow P-C, So C-L, Tu LNL, Roy A, Yap C-W, Kang T-S, et al. Identification of michael acceptor-centric pharmacophores with substituents that yield strong thioredoxin reductase inhibitory character correlated to antiproliferative activity. Antioxid Redox Signal. 2013;19(11):1149–1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zou T, Lum CT, Lok C-N, Zhang J-J, Che C-M.. Chemical biology of anticancer gold (iii) and gold (i) complexes. Chem Soc Rev. 2015;44(24):8786–8801. [DOI] [PubMed] [Google Scholar]
  • 36.Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 2007;7(8):573–584. [DOI] [PubMed] [Google Scholar]
  • 37.Pötsch I, Baier D, Keppler BK, Berger W.. Challenges and chances in the preclinical to clinical translation of anticancer metallodrugs. In: Metal-based Anticancer Agents, ed. Casini, A., Vessières, A., Meier-Menches, S. M. Croydon, UK: The Royal Society of Chemistry, 2019; p. 308–347. [Google Scholar]
  • 38.Giuseppe L, Giuseppa A, Ronga L, Diego T, Ilaria S, Antonina A, Cervello M, Mangiatordi GF, Michele S.. Structure-based identification of the first non-covalent thioredoxin reductase inhibitor with proven ADMET suitability. ChemRxiv. 2025. 10.26434/chemrxiv-2025-xfc4d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cheng Q, Sandalova T, Lindqvist Y, Arnér ESJ.. Crystal structure and catalysis of the selenoprotein thioredoxin reductase 1. J Biol Chem. 2009;284(6):3998–4008. [DOI] [PubMed] [Google Scholar]
  • 40.Halgren TA. Identifying and characterizing binding sites and assessing druggability. J Chem Inf Model. 2009;49(2):377–389. [DOI] [PubMed] [Google Scholar]
  • 41.Madhavi Sastry G, Adzhigirey M, Day T, Annabhimoju R, Sherman W.. Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments. J Comput Aided Mol Des. 2013;27(3):221–234. [DOI] [PubMed] [Google Scholar]
  • 42.Schrödinger Release . Schrödinger Release 2020: Protein Preparation Wizard; Epik; Impact; Prime [software]. New York (NY): Schrödinger, LLC. 2020. [Google Scholar]
  • 43.Schrödinger Release 2020-1: LigPrep. New York, NY: Schrödinger, LLC. 2020. [Google Scholar]
  • 44.Lu C, Wu C, Ghoreishi D, Chen W, Wang L, Damm W, Ross GA, Dahlgren MK, Russell E, Von Bargen CD, et al. OPLS4: Improving force field accuracy on challenging regimes of chemical space. J Chem Theory Comput. 2021;17(7):4291–4300. [DOI] [PubMed] [Google Scholar]
  • 45.Jones G, Willett P, Glen RC, Leach AR, Taylor R.. Development and validation of a genetic algorithm for flexible docking 1 1Edited by F. E. Cohen. J Mol Biol. 1997;267(3):727–748. [DOI] [PubMed] [Google Scholar]
  • 46.Hendlich M, Rippmann F, Barnickel G.. LIGSITE: Automatic and efficient detection of potential small molecule-binding sites in proteins. J Mol Graph Model. 1997;15(6):359–363, 389. [DOI] [PubMed] [Google Scholar]
  • 47.Verdonk ML, Cole JC, Hartshorn MJ, Murray CW, Taylor RD.. Improved protein-ligand docking using GOLD. Proteins Struct. Funct. Bioinforma. 2003;52(4):609–623. [DOI] [PubMed] [Google Scholar]
  • 48.Eldridge MD, Murray CW, Auton TR, Paolini GV, Mee RP.. Empirical scoring functions: I. The development of a fast empirical scoring function to estimate the binding affinity of ligands in receptor complexes. J Comput Aided Mol Des. 1997;11(5):425–445. [DOI] [PubMed] [Google Scholar]
  • 49.Asinex screening libraries . https://www.asinex.com/screening-libraries-(all-libraries.)
  • 50.Fischer A, Smieško M, Sellner M, Lill MA.. Decision making in structure-based drug discovery: Visual inspection of docking results. J Med Chem. 2021;64(5):2489–2500. [DOI] [PubMed] [Google Scholar]
  • 51.Lata S, Akif M.. Structure-based identification of natural compound inhibitor against M. Tuberculosis Thioredoxin Reductase: Insight from molecular docking and dynamics simulation. J Biomol Struct Dyn. 2021;39(12):4480–4489. [DOI] [PubMed] [Google Scholar]
  • 52.Liu R, Shi D, Zhang J, Li X, Han X, Yao X, Fang J.. Virtual screening-guided discovery of thioredoxin reductase inhibitors. Toxicol Appl Pharmacol. 2019;370:106–116. [DOI] [PubMed] [Google Scholar]
  • 53.Li Q, Wall SB, Ren C, Velten M, Hill CL, Locy ML, Rogers LK, Tipple TE.. Thioredoxin reductase inhibition attenuates neonatal hyperoxic lung injury and enhances nuclear factor e2–related factor 2 activation. Am J Respir Cell Mol Biol. 2016;55(3):419–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dunigan K, Li Q, Li R, Locy ML, Wall S, Tipple TE.. The thioredoxin reductase inhibitor auranofin induces heme oxygenase-1 in lung epithelial cells via Nrf2-dependent mechanisms. Am J Physiol Lung Cell Mol Physiol. 2018;315(4):L545–L552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kopp B, Khoury L, Audebert M.. Validation of the γH2AX biomarker for genotoxicity assessment: a review. Arch Toxicol. 2019;93(8):2103–2114. [DOI] [PubMed] [Google Scholar]
  • 56.Zhang S-P, Zhou J, Fan Q-Z, Lv X-M, Wang T, Wang F, Chen Y, Hong S-Y, Liu X-P, Xu B-S, et al. Discovery of hydroxytyrosol as thioredoxin reductase 1 inhibitor to induce apoptosis and g1/s cell cycle arrest in human colorectal cancer cells via ROS generation. Exp Ther Med. 2021;22(2):829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Busker S, Qian W, Haraldsson M, Espinosa B, Johansson L, Attarha S, Kolosenko I, Liu J, Dagnell M, Grandér D, et al. Irreversible TrxR1 inhibitors block STAT3 activity and induce cancer cell death. Sci Adv. 2020;6(12):eaax7945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Davies M, Nowotka M, Papadatos G, Dedman N, Gaulton A, Atkinson F, Bellis L, Overington JP.. ChEMBL web services: streamlining access to drug discovery data and utilities. Nucleic Acids Res. 2015;43(W1):W612–W620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gaulton A, Bellis LJ, Bento AP, Chambers J, Davies M, Hersey A, Light Y, McGlinchey S, Michalovich D, Al-Lazikani B, et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 2012;40(Database issue):D1100–D1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kim S, Chen J, Cheng T, Gindulyte A, He J, He S, Li Q, Shoemaker BA, Thiessen PA, Yu B, et al. PubChem 2023 update. Nucleic Acids Res. 2023;51(D1):D1373–D1380. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary_information_ Clean.docx

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author/s.


Articles from Journal of Enzyme Inhibition and Medicinal Chemistry are provided here courtesy of Taylor & Francis

RESOURCES