Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 263
Filter
1.
J Med Chem ; 67(16): 14543-14552, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39106326

ABSTRACT

Human tryptophan dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO) are two important targets in cancer immunotherapy. Extensive research has led to a large number of potent IDO inhibitors; in addition, 52 structures of IDO in complex with inhibitors with a wide array of chemical scaffolds have been documented. In contrast, progress in the development of TDO inhibitors has been limited. Only four structures of TDO in complex with competitive inhibitors that compete with the substrate L-tryptophan for binding to the active site have been reported to date. Here we systematically evaluated the structures of TDO in complex with competitive inhibitors with three types of pharmacophores, imidazo-isoindole, indole-tetrazole, and indole-benzotriazole. The comparative assessment of the protein-inhibitor interactions sheds new light into the structure-based design of enzyme-selective inhibitors.


Subject(s)
Enzyme Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Tryptophan Oxygenase , Humans , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism , Tryptophan Oxygenase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Structure-Activity Relationship , Indoles/chemistry , Indoles/pharmacology , Indoles/metabolism , Models, Molecular , Tetrazoles/chemistry , Tetrazoles/pharmacology , Tetrazoles/metabolism , Tryptophan/chemistry , Tryptophan/metabolism , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazoles/metabolism , Protein Binding
2.
Comput Biol Med ; 180: 108954, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094327

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) are attractive drug targets for cancer immunotherapy. After disappointing results of the epacadostat as a selective IDO inhibitor in phase III clinical trials, there is much interest in the development of the TDO selective inhibitors. In the current study, several data analysis methods and machine learning approaches including logistic regression, Random Forest, XGBoost and Support Vector Machines were used to model a data set of compounds retrieved from ChEMBL. Models based on the Morgan fingerprints revealed notable fragments for the selective inhibition of the IDO, TDO or both. Multiple fragment docking was performed to find the best set of bound fragments and their orientation in the space for efficient linking. Linking the fragments and optimization of the final molecules were accomplished by means of an artificial intelligence generative framework. Finally, selectivity of the optimized molecules was assessed and the top 4 lead molecules were filtered through PAINS, Brenk and NIH filters. Results indicated that phenyloxalamide, fluoroquinoline, and 3-bromo-4-fluroaniline confer selectivity towards the IDO inhibition. Correspondingly, 1-benzyl-1H-naphtho[2,3-d][1,2,3]triazole-4,9-dione was found to be an integral fragment for the selective inhibition of the TDO by constituting a coordination bond with the Fe atom of heme. In addition, furo[2,3-c]pyridine-2,3-diamine was found as a common fragment for inhibition of the both targets and can be used in the design of the dual target inhibitors of the IDO and TDO. The new fragments introduced here can be a useful building blocks for incorporation into the selective TDO or dual IDO/TDO inhibitors.


Subject(s)
Cheminformatics , Enzyme Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Machine Learning , Tryptophan Oxygenase , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism , Tryptophan Oxygenase/chemistry , Humans , Cheminformatics/methods , Enzyme Inhibitors/chemistry , Molecular Docking Simulation
3.
Front Immunol ; 15: 1440269, 2024.
Article in English | MEDLINE | ID: mdl-39211039

ABSTRACT

Despite the immune system's role in the detection and eradication of abnormal cells, cancer cells often evade elimination by exploitation of various immune escape mechanisms. Among these mechanisms is the ability of cancer cells to upregulate amino acid-metabolizing enzymes, or to induce these enzymes in tumor-infiltrating immunosuppressive cells. Amino acids are fundamental cellular nutrients required for a variety of physiological processes, and their inadequacy can severely impact immune cell function. Amino acid-derived metabolites can additionally dampen the anti-tumor immune response by means of their immunosuppressive activities, whilst some can also promote tumor growth directly. Based on their evident role in tumor immune escape, the amino acid-metabolizing enzymes glutaminase 1 (GLS1), arginase 1 (ARG1), inducible nitric oxide synthase (iNOS), indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO) and interleukin 4 induced 1 (IL4I1) each serve as a promising target for immunotherapeutic intervention. This review summarizes and discusses the involvement of these enzymes in cancer, their effect on the anti-tumor immune response and the recent progress made in the preclinical and clinical evaluation of inhibitors targeting these enzymes.


Subject(s)
Amino Acids , Arginase , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Immunotherapy/methods , Animals , Amino Acids/metabolism , Arginase/metabolism , Glutaminase/metabolism , Glutaminase/antagonists & inhibitors , Tumor Escape , Nitric Oxide Synthase Type II/metabolism , Tryptophan Oxygenase/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Molecular Targeted Therapy , Tumor Microenvironment/immunology , L-Amino Acid Oxidase
4.
BMC Cancer ; 24(1): 889, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048947

ABSTRACT

BACKGROUND: Glioblastoma is an aggressive brain cancer, usually of unknown etiology, and with a very poor prognosis. Survival from diagnosis averages only 3 months if left untreated and this only increases to 12-15 months upon treatment. Treatment options are currently limited and typically comprise radiotherapy plus a course of the DNA-alkylating chemotherapeutic temozolomide. Unfortunately, the disease invariably relapses after several months of treatment with temozolomide, due to the development of resistance to the drug. Increased local tryptophan metabolism is a feature of many solid malignant tumours through increased expression of tryptophan metabolising enzymes. Glioblastomas are notable for featuring increased expression of the tryptophan catabolizing enzymes indole-2,3-dioxygenase-1 (IDO1), and especially tryptophan-2,3-dioxygenase-2 (TDO2). Increased IDO1 and TDO2 activity is known to suppress the cytotoxic T cell response to tumour cells, and this has led to the proposal that the IDO1 and TDO2 enzymes represent promising immuno-oncology targets. In addition to immune modulation, however, recent studies have also identified the activity of these enzymes is important in the development of resistance to chemotherapeutic agents. METHODS: In the current study, the efficacy of a novel dual inhibitor of IDO1 and TDO2, AT-0174, was assessed in an orthotopic mouse model of glioblastoma. C57BL/6J mice were stereotaxically implanted with GL261(luc2) cells into the striatum and then administered either vehicle control, temozolomide (8 mg/kg IP; five 8-day cycles of treatment every 2 days), AT-0174 (120 mg/kg/day PO) or both temozolomide + AT-0174, all given from day 7 after implantation. RESULTS: Temozolomide decreased tumour growth and improved median survival but increased the infiltration of CD4+ Tregs. AT-0174 had no significant effect on tumour growth or survival when given alone, but provided clear synergy in combination with temozolomide, further decreasing tumour growth and significantly improving survival, as well as elevating CD8+ T cell expression and decreasing CD4+ Treg infiltration. CONCLUSION: AT-0174 exhibited an ideal profile for adjunct treatment of glioblastomas with the first-line chemotherapeutic drug temozolomide to prevent development of CD4+ Treg-mediated chemoresistance.


Subject(s)
Drug Synergism , Glioblastoma , Indoleamine-Pyrrole 2,3,-Dioxygenase , Temozolomide , Tryptophan Oxygenase , Animals , Glioblastoma/drug therapy , Glioblastoma/pathology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism , Cell Line, Tumor , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Xenograft Model Antitumor Assays , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use
5.
Phys Chem Chem Phys ; 26(23): 16747-16764, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38818624

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO) is a tryptophan (Trp) metabolic enzyme along the kynurenine (NFK) pathway. Under pathological conditions, IDO overexpressed by tumor cells causes depletion of tryptophan and the accumulation of metabolic products, which inhibit the local immune response and form immune escape. Therefore, the suppression of IDO activity is one of the strategies for tumor immunotherapy, and drug design for this target has been the focus of research for more than two decades. Apart from IDO, tryptophan dioxygenase (TDO) of the same family can also catalyze the same biochemical reaction in the human body, but it has different tissue distribution and substrate selectivity from IDO. Based on the principle of drug design with high potency and low cross-reactivity to specific targets, in this subject, the activity and selectivity of IDO and TDO toward small molecular inhibitors were studied from the perspective of thermodynamics and kinetics. The aim was to elucidate the structural requirements for achieving favorable biological activity and selectivity of IDO and TDO inhibitors. Specifically, the interactions of inhibitors from eight families with IDO and TDO were initially investigated through molecular docking and molecular dynamics simulations, and the thermodynamic data for binding of inhibitors were predicted by the molecular mechanics/generalized Born surface area (MM/GBSA) method. Secondly, we explored the free energy landscape of JKloops, the kinetic control element of IDO/TDO, using temperature replica exchange molecular dynamics (T-REMD) simulations and elucidated the connection between the rules of IDO/TDO conformational changes and the inhibitor selectivity mechanism. Furthermore, the binding and dissociation processes of the C1 inhibitor (NLG919) were simulated by the adaptive steering molecular dynamics (ASMD) method, which not only addressed the possible stable, metastable, and transition states for C1 inhibitor-IDO/TDO interactions, but also accurately predicted kinetic data for C1 inhibitor binding and dissociation. In conclusion, we have constructed a complete process from enzyme (IDO/TDO) conformational activation to inhibitor binding/dissociation and used the thermodynamic and kinetic data of each link as clues to verify the control mechanism of IDO/TDO on inhibitor selectivity. This is of great significance for us to understand the design principles of tumor immunotherapy drugs and to avoid drug resistance of immunotherapy drugs.


Subject(s)
Enzyme Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Thermodynamics , Tryptophan Oxygenase , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Tryptophan Oxygenase/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/chemistry , Molecular Dynamics Simulation , Molecular Docking Simulation , Kinetics
6.
Sci Rep ; 14(1): 9386, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38653790

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.


Subject(s)
Antineoplastic Agents , Drug Repositioning , Indoleamine-Pyrrole 2,3,-Dioxygenase , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism , Cell Line, Tumor , Molecular Docking Simulation , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Drug Screening Assays, Antitumor , Apoptosis/drug effects , Cell Proliferation/drug effects , Pharmacophore
7.
FEBS J ; 291(10): 2172-2190, 2024 May.
Article in English | MEDLINE | ID: mdl-38431776

ABSTRACT

Neuroblastoma poses significant challenges in clinical management. Despite its relatively low incidence, this malignancy contributes disproportionately to cancer-related childhood mortality. Tailoring treatments based on risk stratification, including MYCN oncogene amplification, remains crucial, yet high-risk cases often confront therapeutic resistance and relapse. Here, we explore the aryl hydrocarbon receptor (AHR), a versatile transcription factor implicated in diverse physiological functions such as xenobiotic response, immune modulation, and cell growth. Despite its varying roles in malignancies, AHR's involvement in neuroblastoma remains elusive. Our study investigates the interplay between AHR and its ligand kynurenine (Kyn) in neuroblastoma cells. Kyn is generated from tryptophan (Trp) by the activity of the enzymes indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO2). We found that neuroblastoma cells displayed sensitivity to the TDO2 inhibitor 680C91, exposing potential vulnerabilities. Furthermore, combining TDO2 inhibition with retinoic acid or irinotecan (two chemotherapeutic agents used to treat neuroblastoma patients) revealed synergistic effects in select cell lines. Importantly, clinical correlation analysis using patient data established a link between elevated expression of Kyn-AHR pathway genes and adverse prognosis, particularly in older children. These findings underscore the significance of the Kyn-AHR pathway in neuroblastoma progression, emphasizing its potential role as a therapeutic target.


Subject(s)
Kynurenine , Neuroblastoma , Receptors, Aryl Hydrocarbon , Humans , Kynurenine/metabolism , Neuroblastoma/pathology , Neuroblastoma/metabolism , Neuroblastoma/genetics , Neuroblastoma/drug therapy , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Cell Line, Tumor , Tryptophan Oxygenase/metabolism , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/antagonists & inhibitors , Tretinoin/pharmacology , Signal Transduction/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects
8.
Phytochemistry ; 214: 113794, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37499850

ABSTRACT

The root of Dactylicapnos scandens (D.Don.) Hutch (Papaveraceae), one of the most famous ethno-medicinal plants from the Bai communities in P. R. China, is used to treat various inflammations and tumours. Bioassay-guided phytochemical research on D. scandens followed by semi-synthesis led to a series of undescribed tetrahydroisoquinoline alkaloids with dual inhibitory activities against indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO). The previously undescribed dark-green alkaloid dactycapnine A exhibited the best dual inhibitor effects among the identified compounds. Structure-activity relationship analysis revealed the importance of the base skeleton with a hyperconjugation system. The performed semi-synthesis further yielded bioactive dimeric and trimeric compounds with hyperconjugated systems. Performed STD NMR experiments disclosed direct interactions between dactycapnine A and IDO1/TDO. Inhibition kinetics indicated dactycapnine A as a mixed-type dual inhibitor. These findings provided a possible explanation for the anticancer properties of the ethno-medicinal plant species D. scandens.


Subject(s)
Alkaloids , Antineoplastic Agents , Fumariaceae , Plants, Medicinal , Antineoplastic Agents/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Plants, Medicinal/chemistry , Structure-Activity Relationship , Tryptophan , Tryptophan Oxygenase/antagonists & inhibitors , Fumariaceae/chemistry
9.
Front Immunol ; 13: 807271, 2022.
Article in English | MEDLINE | ID: mdl-35173722

ABSTRACT

Metabolism of tryptophan (Trp), an essential amino acid, represent a major metabolic pathway that both promotes tumor cell intrinsic malignant properties as well as restricts antitumour immunity, thus emerging as a drug development target for cancer immunotherapy. Three cytosolic enzymes, namely indoleamine 2,3-dioxygenase 1 (IDO1), IDO2 and tryptophan 2,3-dioxygenase (TDO2), catalyzes the first-rate limiting step of the degradation of Trp to kynurenine (Kyn) and modulates immunity toward immunosuppression mainly through the aryl hydrocarbon receptor (AhR) activation in numerous types of cancer. By restoring antitumor immune responses and synergizing with other immunotherapies, the encouraging preclinical data of IDO1 inhibitors has dramatically failed to translate into clinical success when combined with immune checkpoints inhibitors, reigniting the debate of combinatorial approach. In this review, we i) provide comprehensive evidences on immunomodulatory role of the Trp catabolism metabolites that highlight this pathway as relevant target in immuno-oncology, ii)ii) discuss underwhelming results from clinical trials investigating efficacy of IDO1 inhibitors and underlying mechanisms that might have contributed to this failure, and finally, iii) discuss the current state-of-art surrounding alternative approaches of innovative antitumor immunotherapies that target molecules of Trp catabolism as well as challenges and perspectives in the era of immunotherapy.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan/metabolism , Animals , Enzyme Inhibitors/therapeutic use , Humans , Immunotherapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan Oxygenase/metabolism
10.
Eur J Med Chem ; 227: 113892, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34678572

ABSTRACT

Selenium is an underexplored element that can be used for bioisosteric replacement of lower molecular weight chalcogens such as oxygen and sulfur. More studies regarding the impact of selenium substitution in different chemical scaffolds are needed to fully grasp this element's potential. Herein, we decided to evaluate the impact of selenium incorporation in a series of tryptophan 2,3-dioxygenase (TDO2) inhibitors, a target of interest in cancer immunotherapy. First, we synthesized the different chalcogen isosteres through Suzuki-Miyaura type coupling. Next, we evaluated the isosteres' affinity and selectivity for TDO2, as well as their lipophilicity, microsomal stability and cellular toxicity on TDO2-expressing cell lines. Overall, chalcogen isosteric replacements did not disturb the on-target activity but allowed for a modulation of the compounds' lipophilicity, toxicity and stability profiles. The present work contributes to our understanding of oxygen/sulfur/selenium isostery towards increasing structural options in medicinal chemistry for the development of novel and distinctive drug candidates.


Subject(s)
Chalcogens/pharmacology , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds/pharmacology , Selenium/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors , Chalcogens/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Humans , Molecular Structure , Oxygen/chemistry , Oxygen/pharmacology , Selenium/chemistry , Stereoisomerism , Structure-Activity Relationship , Sulfur/chemistry , Sulfur/pharmacology , Tryptophan Oxygenase/metabolism
12.
Cancer Lett ; 522: 32-43, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34520819

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzing the conversion of tryptophan (Trp) to kynurenine (Kyn) in kynurenine pathway (KP) is involved in the immunosuppression in pancreatic cancer (PC), but the value of IDO1 as an independent prognostic marker for PC is uncertain. Moreover, the correlation between tryptophan 2,3-dioxygenase (TDO), an isozyme of IDO1, and PC is largely unknown. Using TCGA database, the correlation between IDO1 and/or TDO expression and PC patients' survival was analyzed. The expressions of IDO1 and TDO in PC cells and PC mice were examined. The effects of IDO1, TDO or dual inhibition on IDO1 and TDO effector pathway (Aryl hydrocarbon receptor, AhR) and on migration and invasion of PC cells were investigated. The block effect of IDO1/TDO dual inhibitor RY103 on KP was evaluated. The preclinical efficacy of RY103 and its immunomodulatory effect on KPIC orthotopic PC mice and Pan02 tumor-bearing mice were explored. Results showed that IDO1/TDO co-expression is an independent prognostic marker for PC. RY103 can significantly block KP and target Kyn-AhR pathway to blunt the migration and invasion of PC cells, exhibit preclinical efficacy and ameliorate IDO1/TDO-mediated immunosuppression in PC mice.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Organic Chemicals/pharmacology , Pancreatic Neoplasms/drug therapy , Receptors, Aryl Hydrocarbon/genetics , Tryptophan Oxygenase/genetics , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Kynurenine/biosynthesis , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Organic Chemicals/therapeutic use , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Tryptophan Oxygenase/antagonists & inhibitors , Pancreatic Neoplasms
13.
Biochem Pharmacol ; 193: 114785, 2021 11.
Article in English | MEDLINE | ID: mdl-34562469

ABSTRACT

A tryptophan-2,3-dioxygenase 2 (TDO2)-targeted Pt(IV) prodrug, DN604-TDOi, was designed to prove that the multi-action compound could overcome drug resistance and relieve immunosuppression via introducing a TDO2 inhibitor to the axial position of a six-coordinate Pt(IV) hybrid. Several in vitro biological studies on cisplatin-resistant NSCLC cancer cells suggested that TDO2-targeted Pt(IV) prodrug could combat cisplatin resistance via influencing TDO2-kynurenine (Kyn)-aryl hydrocarbon receptor (AhR)-Aquaporin-4 (AQP4) metabolic circuity and AhR-human DNA polymerase (hpol) κ-induced translesion DNA synthesis (TLS) genomic instability, which are positive in drug-resistant human tumors associated with malignant progression and poor survival. Remarkably, we observed that DN604-TDOi could inhibit TDO2-mediated constitutive Kyn-AhR-AQP4 signaling pathway and suppress hpol κ expression, leading to potential decrease of cell motility and genomic instability in A549/cDDP cells. It was confirmed that TDO2-targeted Pt(IV) prodrug could harness Kyn-AhR-AQP4 metabolic circuitry and TLS genomic instability, exerting antitumor effects in C57BL6 but not TDO2-/- mice. Moreover, the Pt(IV) prodrug improved the intratumoral infiltration of Teff cells and reduced the recruitment of Treg cells. The results provided compelling preclinical evidence that TDO2-targeted Pt(IV) prodrug could abrogate immune chemotherapeutic resistance via decaying TDO2-mediated Kyn-AhR-AQP4 immunosuppression and AhR-hpol κ-induced TLS genomic instability, underscoring the development of a novel Pt(IV)-based candidate as a potent immunotherapeutic agent for chemo-immune resistance prevention.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Tryptophan Oxygenase/antagonists & inhibitors , A549 Cells , Adenocarcinoma of Lung/drug therapy , Carboplatin/analogs & derivatives , Carboplatin/chemistry , Carboplatin/pharmacology , Cell Survival , Cisplatin/pharmacology , Humans , Lung Neoplasms/drug therapy , Platinum Compounds , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/metabolism
14.
J Med Chem ; 64(15): 10967-10980, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34338527

ABSTRACT

Tryptophan 2,3-dioxygenase (TDO2) is a heme-containing enzyme constitutively expressed at high concentrations in the liver and responsible for l-tryptophan (l-Trp) homeostasis. Expression of TDO2 in cancer cells results in the inhibition of immune-mediated tumor rejection due to an enhancement of l-Trp catabolism via the kynurenine pathway. In the study herein, we disclose a new 6-(1H-indol-3-yl)-benzotriazole scaffold of TDO2 inhibitors developed through rational design, starting from existing inhibitors. Rigidification of the initial scaffold led to the synthesis of stable compounds displaying a nanomolar cellular potency and a better understanding of the structural modulations that can be accommodated inside the active site of hTDO2.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Triazoles/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Tryptophan Oxygenase/metabolism , Tumor Cells, Cultured
15.
J Med Chem ; 64(12): 8303-8332, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34110158

ABSTRACT

Human indoleamine 2,3-dioxygenase 1 (hIDO1) and tryptophan 2,3-dioxygenase (hTDO) have been closely linked to the pathogenesis of Parkinson's disease (PD); nevertheless, development of dual hIDO1 and hTDO inhibitors to evaluate their potential efficacy against PD is still lacking. Here, we report biochemical, biophysical, and computational analyses revealing that 1H-indazole-4-amines inhibit both hIDO1 and hTDO by a mechanism involving direct coordination with the heme ferrous and ferric states. Crystal structure-guided optimization led to 23, which manifested IC50 values of 0.64 and 0.04 µM to hIDO1 and hTDO, respectively, and had good pharmacokinetic properties and brain penetration in mice. 23 showed efficacy against the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse motor coordination deficits, comparable to Madopar, an anti-PD medicine. Further studies revealed that different from Madopar, 23 likely has specific anti-PD mechanisms involving lowering IDO1 expression, alleviating dopaminergic neurodegeneration, reducing inflammatory cytokines and quinolinic acid in mouse brain, and increasing kynurenic acid in mouse blood.


Subject(s)
Enzyme Inhibitors/therapeutic use , Indazoles/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/drug therapy , Tryptophan Oxygenase/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/pathology , Cell Line, Tumor , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Indazoles/chemical synthesis , Indazoles/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Protein Binding , Structure-Activity Relationship , Tryptophan Oxygenase/metabolism
16.
ChemMedChem ; 16(18): 2732-2743, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34137184

ABSTRACT

Since its discovery at the beginning of the past century, the essential nutrient l-Tryptophan (l-Trp) and its catabolic pathways have acquired an increasing interest in an ever wider scientific community for their pivotal roles in underlying many important physiological functions and associated pathological conditions. As a consequence, enzymes catalyzing rate limiting steps along l-Trp catabolic pathways - including IDO1, TDO, TPH1 and TPH2 - have turned to be interesting drug targets for the design and development of novel therapeutic agents for different disorders such as carcinoid syndrome, cancer and autoimmune diseases. This article provides a fresh comparative overview on the most recent advancements that crystallographic studies, biophysical and computational works have brought on structural aspects and molecular recognition patterns of these enzymes toward l-Trp. Finally, a conformational analysis of l-Trp is also discussed as part of the molecular recognition process governing the binding of a substrate to its cognate enzymes.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Tryptophan Hydroxylase/antagonists & inhibitors , Tryptophan Oxygenase/antagonists & inhibitors , Binding Sites/drug effects , Enzyme Inhibitors/chemistry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Models, Molecular , Molecular Structure , Tryptophan Hydroxylase/metabolism , Tryptophan Oxygenase/metabolism
17.
Gene ; 792: 145736, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34051337

ABSTRACT

BACKGROUND: The aim of this study was to explore the expression levels and biological significance of TDO2 in colorectal cancer (CRC). METHODS: First, we explored the potential oncogenic roles of TDO2 across 33 tumors based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Second, we evaluated TDO2 protein expression in 55 CRC tissue samples and 30 cDNA samples by immunohistochemistry and qPCR. Third, we investigated the effect of TDO2 on CRC cells by cell proliferation, wound healing, invasion, and colony formation assays. Finally, we determined the protein that is most closely associated with TDO2 via bioinformatics analysis, enriched the key pathways, and verified them. RESULTS: The expression level of TDO2 was found to be associated with the tumor clinical stage in CRC. A high expression of TDO2 was associated with a poor outcome in CRC patients. Inhibition of TDO2 expression by RNAi in LoVo and HCT116 cell lines significantly reduced the proliferation, migration, and invasion abilities as well as colony formation abilities of cells. Further, knockdown of TDO2 expression induced inactivation of the TDO2-KYNU-AhR signaling pathway. CONCLUSION: The results suggest that TDO2 plays an important role in the progression of CRC. Accordingly, TDO2 is a potential therapeutic target in CRC.


Subject(s)
Adenocarcinoma/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Hydrolases/genetics , Receptors, Aryl Hydrocarbon/genetics , Tryptophan Oxygenase/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Atlases as Topic , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Hydrolases/metabolism , Male , Middle Aged , Neoplasm Staging , Prognosis , Protein Binding , Protein Interaction Mapping , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , ROC Curve , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Survival Analysis , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism
18.
Front Immunol ; 12: 636081, 2021.
Article in English | MEDLINE | ID: mdl-33708223

ABSTRACT

Blockade of the immunosuppressive tryptophan catabolism mediated by indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO) holds enormous promise for sensitising cancer patients to immune checkpoint blockade. Yet, only IDO1 inhibitors had entered clinical trials so far, and those agents have generated disappointing clinical results. Improved understanding of molecular mechanisms involved in the immune-regulatory function of the tryptophan catabolism is likely to optimise therapeutic strategies to block this pathway. The immunosuppressive role of tryptophan metabolite kynurenine is becoming increasingly clear, but it remains a mystery if tryptophan exerts functions beyond serving as a precursor for kynurenine. Here we hypothesise that tryptophan acts as a rheostat of kynurenine-mediated immunosuppression by competing with kynurenine for entry into immune T-cells through the amino acid transporter called System L. This hypothesis stems from the observations that elevated tryptophan levels in TDO-knockout mice relieve immunosuppression instigated by IDO1, and that the vacancy of System L transporter modulates kynurenine entry into CD4+ T-cells. This hypothesis has two potential therapeutic implications. Firstly, potent TDO inhibitors are expected to indirectly inhibit IDO1 hence development of TDO-selective inhibitors appears advantageous compared to IDO1-selective and dual IDO1/TDO inhibitors. Secondly, oral supplementation with System L substrates such as leucine represents a novel potential therapeutic modality to restrain the immunosuppressive kynurenine and restore anti-tumour immunity.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Neoplasms/enzymology , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Tumor Escape , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Kynurenine/immunology , Kynurenine/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Tryptophan/immunology , Tryptophan Oxygenase/antagonists & inhibitors , Tumor Escape/drug effects , Tumor Microenvironment
19.
Bioorg Med Chem Lett ; 40: 127910, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33711443

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO1) and tryptophane 2,3-dioxygenase (TDO) are two heme-containing enzymes which catalyze the conversion of tryptophan to N-formylkynurenine. Both enzymes are well establish therapeutic targets as important factors in the tumor immune evasion mechanism. A number of analogues of the marine pyrroloquinoline alkaloids tsitsikammamines or wakayin have been synthesized, two of them were synthesized using an original method to build the bispyrroloquinone framework. All the derivatives were evaluated in a cellular assay for their capacity to inhibit the enzymes. Six compounds have shown a significant potency on HEK 293-EBNA cell lines expressing hIDO1 or hTDO.


Subject(s)
Alkaloids/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Pyrroloiminoquinones/chemical synthesis , Small Molecule Libraries/chemical synthesis , Tryptophan Oxygenase/antagonists & inhibitors , Alkaloids/metabolism , Aquatic Organisms/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Indole Alkaloids/chemistry , Molecular Docking Simulation , Protein Binding , Protein Conformation , Pyrroles/chemistry , Pyrroloiminoquinones/metabolism , Quinolines/chemistry , Small Molecule Libraries/metabolism , Structure-Activity Relationship
20.
Neuropharmacology ; 187: 108490, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33607146

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA) is an amphetamine derivative that has been shown to produce serotonergic damage in the brains of primates, including humans, and of rats. Tryptophan, the precursor of serotonin, is primarily degraded through the kynurenine (KYN) pathway, producing among others KYN, the main metabolite of this route. KYN has been reported as an endogenous agonist of the aryl hydrocarbon receptor (AhR), a transcription factor involved in several neurological functions. This study aims to determine the effect of MDMA on the KYN pathway and on AhR activity and to establish their role in the long-term serotonergic neurotoxicity induced by the drug in rats. Our results show that MDMA induces the activation of the KYN pathway, mediated by hepatic tryptophan 2,3-dioxygenase (TDO). MDMA also activated AhR as evidenced by increased AhR nuclear translocation and CYP1B1 mRNA expression. Autoradiographic quantification of serotonin transporters showed that both the TDO inhibitor 680C91 and the AhR antagonist CH-223191 potentiated the neurotoxicity induced by MDMA, while administration of exogenous l-kynurenine or of the AhR positive modulator 3,3'-diindolylmethane (DIM) partially prevented the serotonergic damage induced by the drug. The results demonstrate for the first time that MDMA increases KYN levels and AhR activity, and these changes appear to play a role in limiting the neurotoxicity induced by the drug. This work provides a better understanding of the physiological mechanisms that attenuate the brain damage induced by MDMA and identify modulation of the KYN pathway and of AhR as potential therapeutic strategies to limit the negative effects of MDMA.


Subject(s)
Hippocampus/drug effects , Kynurenine/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Receptors, Aryl Hydrocarbon/drug effects , Serotonin Agents/toxicity , Tryptophan Oxygenase/drug effects , Animals , Autoradiography , Hippocampus/metabolism , Kynurenine/pharmacology , Neurotoxicity Syndromes , Rats , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Serotonin , Serotonin Plasma Membrane Transport Proteins/metabolism , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL