关键词: Cell cycle arrest Drug repurposing Dual inhibitors Human Indoleamine 2,3-dioxygenase 1 (hIDO1) Human Tryptophan 2,3-Dioxygenase 2 (hTDO2) Pitavastatin

Mesh : Humans Antineoplastic Agents / pharmacology chemistry Indoleamine-Pyrrole 2,3,-Dioxygenase / antagonists & inhibitors metabolism Drug Repositioning Tryptophan Oxygenase / antagonists & inhibitors metabolism Cell Line, Tumor Molecular Docking Simulation Enzyme Inhibitors / pharmacology chemistry Drug Screening Assays, Antitumor Apoptosis / drug effects Cell Proliferation / drug effects Pharmacophore

来  源:   DOI:10.1038/s41598-024-59353-4   PDF(Pubmed)

Abstract:
Discovering effective anti-cancer agents poses a formidable challenge given the limited efficacy of current therapeutic modalities against various cancer types due to intrinsic resistance mechanisms. Cancer immunochemotherapy is an alternative strategy for breast cancer treatment and overcoming cancer resistance. Human Indoleamine 2,3-dioxygenase (hIDO1) and human Tryptophan 2,3-dioxygenase 2 (hTDO2) play pivotal roles in tryptophan metabolism, leading to the generation of kynurenine and other bioactive metabolites. This process facilitates the de novo synthesis of Nicotinamide Dinucleotide (NAD), promoting cancer resistance. This study identified a new dual hIDO1/hTDO2 inhibitor using a drug repurposing strategy of FDA-approved drugs. Herein, we delineate the development of a ligand-based pharmacophore model based on a training set of 12 compounds with reported hIDO1/hTDO2 inhibitory activity. We conducted a pharmacophore search followed by high-throughput virtual screening of 2568 FDA-approved drugs against both enzymes, resulting in ten hits, four of them with high potential of dual inhibitory activity. For further in silico and in vitro biological investigation, the anti-hypercholesterolemic drug Pitavastatin deemed the drug of choice in this study. Molecular dynamics (MD) simulations demonstrated that Pitavastatin forms stable complexes with both hIDO1 and hTDO2 receptors, providing a structural basis for its potential therapeutic efficacy. At nanomolar (nM) concentration, it exhibited remarkable in vitro enzyme inhibitory activity against both examined enzymes. Additionally, Pitavastatin demonstrated potent cytotoxic activity against BT-549, MCF-7, and HepG2 cell lines (IC50 = 16.82, 9.52, and 1.84 µM, respectively). Its anticancer activity was primarily due to the induction of G1/S phase arrest as discovered through cell cycle analysis of HepG2 cancer cells. Ultimately, treating HepG2 cancer cells with Pitavastatin affected significant activation of caspase-3 accompanied by down-regulation of cellular apoptotic biomarkers such as IDO, TDO, STAT3, P21, P27, IL-6, and AhR.
摘要:
鉴于由于固有的抗癌性机制而导致的针对各种癌症类型的当前治疗方式的有限功效,发现有效的抗癌剂提出了巨大的挑战。癌症免疫化疗是乳腺癌治疗和克服癌症耐药性的替代策略。人吲哚胺2,3-双加氧酶(hIDO1)和人色氨酸2,3-双加氧酶2(hTDO2)在色氨酸代谢中起关键作用,导致犬尿氨酸和其他生物活性代谢产物的产生。此过程促进烟酰胺二核苷酸(NAD)的从头合成,促进抗癌性。这项研究使用FDA批准的药物的药物再利用策略确定了一种新的双重hIDO1/hTDO2抑制剂。在这里,我们描述了基于配体的药效团模型的发展,该模型基于12种已报道hIDO1/hTDO2抑制活性的化合物的训练集。我们进行了药效团搜索,然后对2568种FDA批准的针对这两种酶的药物进行了高通量虚拟筛选。导致十次点击,其中四个具有双重抑制活性的高潜力。为了进一步进行计算机模拟和体外生物学研究,抗高胆固醇血症药物匹伐他汀被认为是本研究的首选药物。分子动力学(MD)模拟表明,匹伐他汀与hIDO1和hTDO2受体形成稳定的复合物,为其潜在的治疗功效提供结构基础。在纳摩尔(nM)浓度下,它对两种检查的酶都表现出显着的体外酶抑制活性。此外,匹伐他汀对BT-549,MCF-7和HepG2细胞系表现出有效的细胞毒性活性(IC50=16.82,9.52和1.84µM,分别)。其抗癌活性主要是由于通过HepG2癌细胞的细胞周期分析发现的G1/S期停滞的诱导。最终,用匹伐他汀治疗HepG2癌细胞影响caspase-3的显著激活,伴随着细胞凋亡生物标志物如IDO的下调,TDO,STAT3、P21、P27、IL-6和AhR。
公众号