1
|
Wang T, Liao X, Zhao X, Chen K, Chen Y, Wen H, Yin D, Wang Y, Lin B, Zhang S, Cui H. Rational design of 2-benzylsulfinyl-benzoxazoles as potent and selective indoleamine 2,3-dioxygenase 1 inhibitors to combat inflammation. Bioorg Chem 2024; 152:107740. [PMID: 39217780 DOI: 10.1016/j.bioorg.2024.107740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Mimicking the transition state of tryptophan (Trp) and O2 in the enzymatic reaction is an effective approach to design indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. In this study, we firstly assembled a small library of 2-substituted benzo-fused five membered heterocycles and found 2-sulfinyl-benzoxazoles with interesting IDO1 inhibitory activities. Next the inhibitory activity toward IDO1 was gradually improved. Several benzoxazoles showed potent IDO1 inhibitory activity with IC50 of 82-91 nM, and exhibited selectivity between IDO1 and tryptophan 2,3-dioxygenase (TDO2). Enzyme binding studies showed that benzoxazoles are reversible type II IDO1 inhibitors, and modeling studies suggested that the oxygen atom of the sulfoxide in benzoxazoles interacts with the iron atom of the heme group, which mimics the transition state of Fe-O-O-Trp complex. Especially, 10b can effectively inhibit the NO production in lipopolysaccharides (LPS) stimulated RAW264.7 cells, and it also shows good anti-inflammation effect on mice acute inflammation model of croton oil induced ear edema.
Collapse
Affiliation(s)
- Ting Wang
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Xiufeng Liao
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Xiaodi Zhao
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Kai Chen
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yangzhonghui Chen
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Hui Wen
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Dali Yin
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Yuchen Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China.
| | - Bin Lin
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Sen Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China.
| | - Huaqing Cui
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China.
| |
Collapse
|
2
|
Hennes E, Lucas B, Scholes NS, Cheng XF, Scott DC, Bischoff M, Reich K, Gasper R, Lucas M, Xu TT, Pulvermacher LM, Dötsch L, Imrichova H, Brause A, Naredla KR, Sievers S, Kumar K, Janning P, Gersch M, Murray PJ, Schulman BA, Winter GE, Ziegler S, Waldmann H. Monovalent Pseudo-Natural Product Degraders Supercharge the Native Degradation of IDO1 by KLHDC3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602857. [PMID: 39026748 PMCID: PMC11257573 DOI: 10.1101/2024.07.10.602857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Targeted protein degradation (TPD) modulates protein function beyond inhibition of enzyme activity or protein-protein interactions. Most degraders function by proximity induction, and directly bridge an E3 ligase with the target to be degraded. However, many proteins might not be addressable via proximity-based degraders, and other challenges, such as resistance acquisition, exist. Here, we identified pseudo-natural products derived from (-)-myrtanol, termed iDegs, that inhibit and induce degradation of the immunomodulatory enzyme indoleamine-2,3-dioxygenase 1 (IDO1) by a distinct mechanism. iDegs induce a unique conformational change and, thereby, boost IDO1 ubiquitination and degradation by the cullin-RING E3 ligase CRL2KLHDC3, which we identified to also mediate native IDO1 degradation. Therefore, iDegs supercharge the native proteolytic pathway of IDO1, rendering this mechanism of action distinct from traditional degrader approaches involving proteolysis-targeting chimeras (PROTACs) or molecular-glue degraders (MGDs). In contrast to clinically explored IDO1 inhibitors, iDegs reduce formation of kynurenine by both inhibition and induced degradation of the enzyme and should also modulate non-enzymatic functions of IDO1. This unique mechanism of action may open up new therapeutic opportunities for the treatment of cancer beyond classical inhibition of IDO1.
Collapse
Affiliation(s)
- Elisabeth Hennes
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
| | - Belén Lucas
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Natalie S. Scholes
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Xiu-Fen Cheng
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Daniel C. Scott
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Matthias Bischoff
- Compound Management and Screening Center Otto-Hahn-Str.11, 44227 Dortmund, Germany
| | - Katharina Reich
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Raphael Gasper
- Max-Planck-Institut für Molekulare Physiologie, Zentrale Einheit für Kristallographie und Biophysik, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - María Lucas
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, C/Albert Einstein 22, PCTCAN, 39011 Santander, Spain
| | - Teng Teng Xu
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Lisa-Marie Pulvermacher
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Lara Dötsch
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
| | - Hana Imrichova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alexandra Brause
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Kesava Reddy Naredla
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Sonja Sievers
- Compound Management and Screening Center Otto-Hahn-Str.11, 44227 Dortmund, Germany
| | - Kamal Kumar
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Petra Janning
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Malte Gersch
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
- Chemical Genomics Centre, Max-Planck-Institut für Molekulare Physiologie, 44227 Dortmund, Germany
| | - Peter J. Murray
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Brenda A. Schulman
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Georg E. Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Slava Ziegler
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
| |
Collapse
|
3
|
Mirgaux M, Leherte L, Wouters J. Human indoleamine-2,3-dioxygenase 2 cofactor lability and low substrate affinity explained by homology modeling, molecular dynamics and molecular docking. J Biomol Struct Dyn 2024; 42:4475-4488. [PMID: 37301605 DOI: 10.1080/07391102.2023.2220830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023]
Abstract
The human indoleamine-2,3-dioxygenase 2 (hIDO2) protein is growing of interest as it is increasingly implicated in multiple diseases (cancer, autoimmune diseases, COVID-19). However, it is only poorly reported in the literature. Its mode of action remains unknown because it does not seem to catalyze the reaction for which it is attributed: the degradation of the L-Tryptophan into N-formyl-kynurenine. This contrasts with its paralog, the human indoleamine-2,3-dioxygenase 1 (hIDO1), which has been extensively studied in the literature and for which several inhibitors are already in clinical trials. Yet, the recent failure of one of the most advanced hIDO1 inhibitors, the Epacadostat, could be caused by a still unknown interaction between hIDO1 and hIDO2. In order to better understand the mechanism of hIDO2, and in the absence of experimental structural data, a computational study mixing homology modeling, Molecular Dynamics, and molecular docking was conducted. The present article highlights an exacerbated lability of the cofactor as well as an inadequate positioning of the substrate in the active site of hIDO2, which might bring part of an answer to its lack of activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manon Mirgaux
- Department of Chemistry, Laboratoire de Chimie Biologique Structurale (CBS), Namur Institute of Structured Matter (NISM), Namur Research Institute for Life Science (NARILIS), University of Namur (UNamur), Namur, Belgium
| | - Laurence Leherte
- Department of Chemistry, Laboratoire de Chimie Biologique Structurale (CBS), Namur Institute of Structured Matter (NISM), Namur Research Institute for Life Science (NARILIS), University of Namur (UNamur), Namur, Belgium
| | - Johan Wouters
- Department of Chemistry, Laboratoire de Chimie Biologique Structurale (CBS), Namur Institute of Structured Matter (NISM), Namur Research Institute for Life Science (NARILIS), University of Namur (UNamur), Namur, Belgium
| |
Collapse
|
4
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
5
|
Rossini S, Ambrosino S, Volpi C, Belladonna ML, Pallotta MT, Panfili E, Suvieri C, Macchiarulo A, Mondanelli G, Orabona C. Epacadostat stabilizes the apo-form of IDO1 and signals a pro-tumorigenic pathway in human ovarian cancer cells. Front Immunol 2024; 15:1346686. [PMID: 38333210 PMCID: PMC10850306 DOI: 10.3389/fimmu.2024.1346686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
The tryptophan-degrading enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is a plastic immune checkpoint molecule that potently orchestrates immune responses within the tumor microenvironment (TME). As a heme-containing protein, IDO1 catalyzes the conversion of the essential amino acid tryptophan into immunoactive metabolites, called kynurenines. By depleting tryptophan and enriching the TME with kynurenines, IDO1 catalytic activity shapes an immunosuppressive TME. Accordingly, the inducible or constitutive IDO1 expression in cancer correlates with a negative prognosis for patients, representing one of the critical tumor-escape mechanisms. However, clinically trialed IDO1 catalytic inhibitors disappointed the expected anti-tumor efficacy. Interestingly, the non-enzymatic apo-form of IDO1 is still active as a transducing protein, capable of promoting an immunoregulatory phenotype in dendritic cells (DCs) as well as a pro-tumorigenic behavior in murine melanoma. Moreover, the IDO1 catalytic inhibitor epacadostat can induce a tolerogenic phenotype in plasmacytoid DCs, overcoming the catalytic inhibition of IDO1. Based on this recent evidence, IDO1 plasticity was investigated in the human ovarian cancer cell line, SKOV-3, that constitutively expresses IDO1 in a dynamic balance between the holo- and apo-protein, and thus potentially endowed with a dual function (i.e., enzymatic and non-enzymatic). Besides inhibiting the catalytic activity, epacadostat persistently stabilizes the apo-form of IDO1 protein, favoring its tyrosine-phosphorylation and promoting its association with the phosphatase SHP-2. In SKOV-3 cells, both these early molecular events activate a signaling pathway transduced by IDO1 apo-protein, which is independent of its catalytic activity and contributes to the tumorigenic phenotype of SKOV-3 cells. Overall, our findings unveiled a new mechanism of action of epacadostat on IDO1 target, repositioning the catalytic inhibitor as a stabilizer of the apo-form of IDO1, still capable of transducing a pro-tumorigenic pathway in SKOV-3 tumor. This mechanism could contribute to clarify the lack of effectiveness of epacadostat in clinical trials and shed light on innovative immunotherapeutic strategies to tackle IDO1 target.
Collapse
Affiliation(s)
- Sofia Rossini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sara Ambrosino
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudia Volpi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | - Eleonora Panfili
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Chiara Suvieri
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giada Mondanelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ciriana Orabona
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
6
|
Singh R, Kumar R, Roy A, Behera PM, Atri AK, Kumar K, Manna D, Dixit A, Patil MT, Mankamna Kumari R, Nimesh S, Salunke DB. Imidazo[2,1-b]thiazole based indoleamine-2,3-dioxygenase 1 (IDO1) inhibitor: Structure based design, synthesis, bio-evaluation and docking studies. Bioorg Med Chem Lett 2023; 96:129532. [PMID: 37866714 DOI: 10.1016/j.bmcl.2023.129532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/25/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Indoleamine-2,3-dioxygenase 1 (IDO1) is an immunomodulatory enzyme known to catalyse the initial and rate limiting step of kynurenine pathway of l-tryptophan metabolism. IDO1 enzyme over expression plays a crucial role in progression of cancer, malaria, multiple sclerosis and other life-threatening diseases. Several efforts over the last two decades have been invested by the researchers for the discovery of different IDO1 inhibitors and the plasticity of the IDO1 enzyme ligand binding pocket provide ample opportunities to develop new heterocyclic scaffolds targeting this enzyme. In the present work, based on the X-ray crystal structure of human IDO1 coordinated with few ligands, we designed and synthesized new fused heterocyclic compounds and evaluated their potential human IDO1 inhibitory activity (compound 30 and 41 showed IC50 values of 23 and 13 µM, respectively). The identified HITs were observed to be non-toxic to HEK293 cells at 100 µM concentration. The observed activity of the synthesized compounds was correlated with the specific interactions of their structures at the enzyme pocket using docking studies. A detailed analysis of docking results of the synthesized analogues as well as selected known IDO1 inhibitors revealed that most of the inhibitors have some reasonable docking scores in at least two crystal structures and have similar orientation as that of co-crystal ligands.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Ravinder Kumar
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Ashalata Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Pabitra Mohan Behera
- Institute of Life Science, Nalco Square, Chandrasekharpur, Bhubaneswar 751023, India
| | - Ankit K Atri
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Kushvinder Kumar
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Debasis Manna
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Anshuman Dixit
- Institute of Life Science, Nalco Square, Chandrasekharpur, Bhubaneswar 751023, India
| | - Madhuri T Patil
- Department of Chemistry, Mehr Chand Mahajan DAV College for Women, Chandigarh 160 036, India
| | - R Mankamna Kumari
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Ajmer 305 801, India
| | - Surendra Nimesh
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Ajmer 305 801, India
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India; National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
7
|
Sun H, Yang Q, Yu X, Huang M, Ding M, Li W, Tang Y, Liu G. Prediction of IDO1 Inhibitors by a Fingerprint-Based Stacking Ensemble Model Named IDO1Stack. ChemMedChem 2023; 18:e202300151. [PMID: 37340939 DOI: 10.1002/cmdc.202300151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/22/2023]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is viewed as an extremely promising target for cancer immunotherapy. Here, we proposed a two-layer stacking ensemble model, IDO1Stack, that can efficiently predict IDO1 inhibitors. First, we constructed a series of classification models based on five machine learning algorithms and eight molecular characterization methods. Then, a stacking ensemble model was built using the top five models as the base classifier and logistic regression as the meta-classifier. The areas under the receiver operating characteristic curve (AUC) of IDO1Stack on the test set and external validation set were 0.952 and 0.918, respectively. Furthermore, we computed the applicability domain and privileged substructures of the model and interpreted the model using SHapley Additive exPlanations (SHAP). It is expected that IDO1Stack can well study the interaction between target and ligand, providing practitioners with a reliable tool for rapid screening and discovery of IDO1 inhibitors.
Collapse
Affiliation(s)
- Huimin Sun
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Qing Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Xinxin Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Mengting Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Meng Ding
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
8
|
Hoang VH, Trang NTK, Minh TC, Long LTB, Lan TH, Hue NT, Tien LQ, Nguyen TX, Nguyen YTK, Yoo H, Tran PT. Design, synthesis and evaluation the bioactivities of novel 1,3-dimethyl-6-amino-1H-indazole derivatives as anticancer agents. Bioorg Med Chem 2023; 90:117377. [PMID: 37352576 DOI: 10.1016/j.bmc.2023.117377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/25/2023]
Abstract
Indoleamine 2,3-dioxygenase (IDO1) is a heme-containing enzyme mainly responsible for the metabolism of tryptophan to kynurenine. To date, the IDO1 inhibitors have been developed intensively for the re-activation of the anticancer immune response. In this report, we designed, and synthesized novel 1,3-dimethyl-6-amino indazole derivatives as IDO1 inhibitors based on the structure of IDO1 active site. We further examined their anticancer activity on hypopharyngeal carcinoma cells (FaDu), squamous cell carcinoma of the oral tongue (YD-15), breast cancer cells (MCF7), and human dental pulp stem cells (HDPSC). Of them, compound N-(4-bromobenzyl)-1,3-dimethyl-1H-indazol-6-amine (7) remarkably suppressed IDO1 expression in a concentration - dependent manner. In addition, 7 was the most potential anticancer compound with inducing apoptosis activity as well as selectively activated extracellular signal-regulated kinases (ERK) in mitogen-activated protein kinase (MAPK) pathways on FaDu cells. Finally, compound 7 suppressed cell mobility in wound healing assay with the reduced expression of matrix metalloproteinase MMP9. Taken together, we believe that 7 is the most promising compound, which may be applied to treatment of hypopharyngeal carcinoma.
Collapse
Affiliation(s)
- Van-Hai Hoang
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Viet Nam; PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Yen Nghia, Hadong, Hanoi 12116, Viet Nam
| | - Nguyen Thi Kieu Trang
- Department of Pharmacology and Dental Therapeutics, College of Dentistry, Chosun University, Gwangju 61452, South Korea; Department of Pharmacy, Thai Binh University of Medicine and Pharmacy, Thai Binh City 06000, Viet Nam
| | - Truong Cao Minh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Le Thien Bao Long
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Tran Hoang Lan
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Nguyen Thi Hue
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Le Quoc Tien
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Thanh Xuan Nguyen
- Department of Neurosurgery, Viet-Duc University Hospital, Hanoi 100000, Viet Nam
| | - Yen Thi Kim Nguyen
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hoon Yoo
- Department of Pharmacology and Dental Therapeutics, College of Dentistry, Chosun University, Gwangju 61452, South Korea.
| | - Phuong-Thao Tran
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam.
| |
Collapse
|
9
|
Savonije K, Meek A, Weaver DF. Indoleamine 2,3-Dioxygenase as a Therapeutic Target for Alzheimer's Disease and Geriatric Depression. Brain Sci 2023; 13:852. [PMID: 37371332 DOI: 10.3390/brainsci13060852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Neuroimmune-triggered neuroinflammation of the central nervous system is emerging as an important aetiopathogenic factor for multiple neurological disorders, including depression, dementia, Alzheimer's disease, multiple sclerosis and others. Tryptophan metabolism via the kynurenic pathway, which is initiated by the indoleamine-2,3-dioxygenase (IDO-1) enzyme, is a key regulator of the neuroimmune system and its associated neuroinflammatory effects. As discussed in this review, targeting the production of immunopathic and potentially neurotoxic kynurenine metabolites by inhibitory downregulation of IDO-1 may prove a viable target against inflammation-induced neurological conditions, particularly depression and dementia.
Collapse
Affiliation(s)
- Karl Savonije
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Autumn Meek
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Donald F Weaver
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Departments of Chemistry and Medicine, University of Toronto, Toronto, ON M5S 3M2, Canada
| |
Collapse
|
10
|
Markwalder JA, Balog AJ, Williams DK, Nara SJ, Reddy R, Roy S, Kanyaboina Y, Li X, Johnston K, Fan Y, Lewis H, Marsilio F, Yan C, Critton D, Newitt JA, Traeger SC, Wu DR, Jure-Kunkel MN, Jayaraman L, Lin TA, Sinz MW, Hunt JT, Seitz SP. Synthesis and Biological Evaluation of Biaryl Alkyl Ethers as Inhibitors of IDO1. Bioorg Med Chem Lett 2023; 88:129280. [PMID: 37054759 DOI: 10.1016/j.bmcl.2023.129280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/15/2023]
Abstract
Starting from the dialkylaniline indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor lead 3 (IDO1 HeLa IC50 = 7.0 nM), an iterative process of synthesis and screening led to cyclized analog 21 (IDO1 HeLa IC50 = 3.6 nM) which maintained the high potency of 3 while addressing issues of lipophilicity, cytochrome P450 (CYP) inhibition, hERG (human potassium ion channel Kv11.1) inhibition, Pregnane X Receptor (PXR) transactivation, and oxidative metabolic stability. An x-ray crystal structure of a biaryl alkyl ether 11 bound to IDO1 was obtained. Consistent with our earlier results, compound 11 was shown to bind to the apo form of the enzyme.
Collapse
Affiliation(s)
- Jay A Markwalder
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States.
| | - Aaron J Balog
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | - David K Williams
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States.
| | - Susheel J Nara
- Discovery Chemistry, Biocon Bristol Myers Squibb Research & Development Center, Bangalore, India
| | - Ratnakar Reddy
- Discovery Chemistry, Biocon Bristol Myers Squibb Research & Development Center, Bangalore, India
| | - Saumya Roy
- Former Bristol Myers Squibb Employee, USA
| | - Yadagiri Kanyaboina
- Discovery Chemistry, Biocon Bristol Myers Squibb Research & Development Center, Bangalore, India
| | - Xin Li
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | | | - Yi Fan
- Former Bristol Myers Squibb Employee, USA
| | - Hal Lewis
- Former Bristol Myers Squibb Employee, USA
| | - Frank Marsilio
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | - Chunhong Yan
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | - David Critton
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | - John A Newitt
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | - Sarah C Traeger
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | - Dauh-Rurng Wu
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | | | | | - Tai-An Lin
- Former Bristol Myers Squibb Employee, USA
| | - Michael W Sinz
- Research & Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, NJ 08543, United States
| | | | | |
Collapse
|
11
|
Röhrig UF, Majjigapu SR, Vogel P, Reynaud A, Pojer F, Dilek N, Reichenbach P, Ascenção K, Irving M, Coukos G, Michielin O, Zoete V. Structure-based optimization of type III indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. J Enzyme Inhib Med Chem 2022; 37:1773-1811. [PMID: 35758198 PMCID: PMC9246256 DOI: 10.1080/14756366.2022.2089665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The haem enzyme indoleamine 2,3-dioxygenase 1 (IDO1) catalyses the rate-limiting step in the kynurenine pathway of tryptophan metabolism and plays an essential role in immunity, neuronal function, and ageing. Expression of IDO1 in cancer cells results in the suppression of an immune response, and therefore IDO1 inhibitors have been developed for use in anti-cancer immunotherapy. Here, we report an extension of our previously described highly efficient haem-binding 1,2,3-triazole and 1,2,4-triazole inhibitor series, the best compound having both enzymatic and cellular IC50 values of 34 nM. We provide enzymatic inhibition data for almost 100 new compounds and X-ray diffraction data for one compound in complex with IDO1. Structural and computational studies explain the dramatic drop in activity upon extension to pocket B, which has been observed in diverse haem-binding inhibitor scaffolds. Our data provides important insights for future IDO1 inhibitor design.
Collapse
Affiliation(s)
- Ute F Röhrig
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Lausanne, Switzerland
| | - Somi Reddy Majjigapu
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Lausanne, Switzerland.,Laboratory of Glycochemistry and Asymmetric Synthesis, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pierre Vogel
- Laboratory of Glycochemistry and Asymmetric Synthesis, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Aline Reynaud
- Protein Production and Structure Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Florence Pojer
- Protein Production and Structure Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nahzli Dilek
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Lausanne, Switzerland
| | - Patrick Reichenbach
- Department of Oncology UNIL-CHUV, Ludwig Lausanne Branch, Epalinges, Switzerland
| | - Kelly Ascenção
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology UNIL-CHUV, Ludwig Lausanne Branch, Epalinges, Switzerland
| | - George Coukos
- Department of Oncology UNIL-CHUV, Ludwig Lausanne Branch, Epalinges, Switzerland
| | - Olivier Michielin
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne (CHUV), Ludwig Cancer Research-Lausanne Branch, Lausanne, CH-1011, Switzerland
| | - Vincent Zoete
- SIB Swiss Institute of Bioinformatics, Molecular Modeling Group, Lausanne, Switzerland.,Department of Oncology UNIL-CHUV, Ludwig Lausanne Branch, Epalinges, Switzerland
| |
Collapse
|
12
|
Davies C, Dötsch L, Ciulla MG, Hennes E, Yoshida K, Gasper R, Scheel R, Sievers S, Strohmann C, Kumar K, Ziegler S, Waldmann H. Identification of a Novel Pseudo-Natural Product Type IV IDO1 Inhibitor Chemotype. Angew Chem Int Ed Engl 2022; 61:e202209374. [PMID: 35959923 DOI: 10.1002/anie.202209374] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Indexed: 01/07/2023]
Abstract
Natural product (NP)-inspired design principles provide invaluable guidance for bioactive compound discovery. Pseudo-natural products (PNPs) are de novo combinations of NP fragments to target biologically relevant chemical space not covered by NPs. We describe the design and synthesis of apoxidoles, a novel pseudo-NP class, whereby indole- and tetrahydropyridine fragments are linked in monopodal connectivity not found in nature. Apoxidoles are efficiently accessible by an enantioselective [4+2] annulation reaction. Biological evaluation revealed that apoxidoles define a new potent type IV inhibitor chemotype of indoleamine 2,3-dioxygenase 1 (IDO1), a heme-containing enzyme considered a target for the treatment of neurodegeneration, autoimmunity and cancer. Apoxidoles target apo-IDO1, prevent heme binding and induce unique amino acid positioning as revealed by crystal structure analysis. Novel type IV apo-IDO1 inhibitors are in high demand, and apoxidoles may provide new opportunities for chemical biology and medicinal chemistry research.
Collapse
Affiliation(s)
- Caitlin Davies
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Lara Dötsch
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Maria Gessica Ciulla
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Current address: Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Elisabeth Hennes
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Kei Yoshida
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Raphael Gasper
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Rebecca Scheel
- Technical University of Dortmund, Department of Inorganic Chemistry, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Sonja Sievers
- Compound Management and Screening Center (COMAS), Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Carsten Strohmann
- Technical University of Dortmund, Department of Inorganic Chemistry, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Kamal Kumar
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Current address: AiCuris Anti-infective Cures AG, Friedrich-Ebert-Str. 475, 42117, Wuppertal, Germany
| | - Slava Ziegler
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Herbert Waldmann
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| |
Collapse
|
13
|
Davies C, Dötsch L, Ciulla MG, Hennes E, Yoshida K, Gasper R, Scheel R, Sievers S, Strohmann C, Kumar K, Ziegler S, Waldmann H. Identification of a Novel Pseudo‐Natural Product Type IV IDO1 Inhibitor Chemotype. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Caitlin Davies
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Lara Dötsch
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Maria Gessica Ciulla
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Elisabeth Hennes
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Kei Yoshida
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Raphael Gasper
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Crystallography and Biophysics Facility GERMANY
| | - Rebecca Scheel
- Technische Universität Dortmund: Technische Universitat Dortmund Inorganic Chemistry GERMANY
| | - Sonja Sievers
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Compound Management and Screening Center GERMANY
| | - Carsten Strohmann
- Technische Universität Dortmund: Technische Universitat Dortmund Inorganic Chemistry GERMANY
| | - Kamal Kumar
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Slava Ziegler
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Herbert Waldmann
- Max-Planck-Institute of Molecular Physiology: Max-Planck-Institut fur molekulare Physiologie Chemical Biology Otto-Hahn-Str. 11 44227 Dortmund GERMANY
| |
Collapse
|
14
|
Dual-target inhibitors of indoleamine 2, 3 dioxygenase 1 (Ido1): A promising direction in cancer immunotherapy. Eur J Med Chem 2022; 238:114524. [PMID: 35696861 DOI: 10.1016/j.ejmech.2022.114524] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023]
Abstract
Indoleamine 2, 3-dioxygenase 1 (IDO1) is a rate-limiting enzyme that catalyzes the kynurenine (Kyn) pathway of tryptophan metabolism in the first step, and the kynurenine pathway plays a fundamental role in immunosuppression in the tumor microenvironment. Therefore, researchers are vigorously developing IDO1 inhibitors, hoping to apply them to cancer immunotherapy. Nowadays, there have been 11 kinds of IDO1 inhibitors entering clinical trials, among which many inhibitors have shown good tumor inhibitory effect in phase I/II clinical trials. But the phase III study of the most promising IDO1 inhibitor compound 29 (Epacadostat) failed in 2018, which may be caused by the compensation effect offered by tryptophan 2,3-dioxygenase (TDO), the mismatched drug combination strategies, or other reasons. Luckily, dual-target inhibitors show great potential and advantages in solving these problems. In recent years, many studies have linked IDO1 to popular targets and selected many IDO1 dual-target inhibitors through pharmacophore fusion strategy and library construction, which enhance the tumor inhibitory effect and reduce side effects. Currently, three kinds of IDO1/TDO dual-target inhibitors have entered clinical trials, and extensive studies have been developing on IDO1 dual-target inhibitors. In this review, we summarize the IDO1 dual-target inhibitors developed in recent years and focus on the structure optimization process, structure-activity relationship, and the efficacy of in vitro and in vivo experiments, shedding a light on the pivotal significance of IDO1 dual-target inhibitors in the treatment of cancer, providing inspiration for the development of new IDO1 dual-target inhibitors.
Collapse
|
15
|
Mammoli A, Bianconi E, Ruta L, Riccio A, Bigiotti C, Souma M, Carotti A, Rossini S, Suvieri C, Pallotta MT, Grohmann U, Camaioni E, Macchiarulo A. Critical Assessment of a Structure-Based Screening Campaign for IDO1 Inhibitors: Tips and Pitfalls. Int J Mol Sci 2022; 23:ijms23073981. [PMID: 35409342 PMCID: PMC8999677 DOI: 10.3390/ijms23073981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/26/2022] Open
Abstract
Over the last two decades, indoleamine 2,3-dioxygenase 1 (IDO1) has attracted wide interest as a key player in immune regulation, fostering the design and development of small molecule inhibitors to restore immune response in tumor immunity. In this framework, biochemical, structural, and pharmacological studies have unveiled peculiar structural plasticity of IDO1, with different conformations and functional states that are coupled to fine regulation of its catalytic activity and non-enzymic functions. The large plasticity of IDO1 may affect its ligand recognition process, generating bias in structure-based drug design campaigns. In this work, we report a screening campaign of a fragment library of compounds, grounding on the use of three distinct conformations of IDO1 that recapitulate its structural plasticity to some extent. Results are instrumental to discuss tips and pitfalls that, due to the large plasticity of the enzyme, may influence the identification of novel and differentiated chemical scaffolds of IDO1 ligands in structure-based screening campaigns.
Collapse
Affiliation(s)
- Andrea Mammoli
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Luana Ruta
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Alessandra Riccio
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Carlo Bigiotti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Maria Souma
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Andrea Carotti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Sofia Rossini
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Chiara Suvieri
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Maria Teresa Pallotta
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Ursula Grohmann
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Emidio Camaioni
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
- Correspondence: ; Tel.: +39-(075)-585-5131
| |
Collapse
|