1
|
Vinogradova L, Komarova K, Lukin A, Zhuravlev M, Deniskin D, Poliakova A, Chudinov M, Gureev M, Dogonadze M, Vinogradova T, Rogacheva E, Kraeva L, Porozov Y, Korzhikov-Vlakh V. You Win Some, You Lose Some: Modifying the Molecular Periphery of Nitrofuran-Tagged Diazaspirooctane Reshapes Its Antibacterial Activity Profile. Int J Mol Sci 2024; 26:207. [PMID: 39796065 PMCID: PMC11720470 DOI: 10.3390/ijms26010207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
The use of the concept of privileged structures significantly accelerates the search for new leads and their optimization. 6-(methylsulfonyl)-8-(4-methyl-4H-1,2,4-triazol-3-yl)-2-(5-nitro-2-furoyl)-2,6-diazaspiro[3.4]octane 1 has been identified as a lead, with MICs of 0.0124-0.0441 μg/mL against MTb multiresistant strains. Several series of structural analogues have been synthesized, including variations in the periphery and simplifications of their scaffolds. All synthesized compounds were tested against the MTb H37Rv strain and ESKAPE panel of pathogens using serial broth dilutions. However, an attempt to optimize structure of 1 did not lead to the development of more active compounds which can work against MTb, but to substances with high activity against S. aureus. Induced-fit docking and MM-GBSA calculations determined a change in the likely biotarget from deazaflavin-dependent nitroreductase to azoreductases. The privileged nature of the scaffold was demonstrated by the detection of a different type of activity.
Collapse
Affiliation(s)
- Lyubov Vinogradova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Kristina Komarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Alexey Lukin
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Maxim Zhuravlev
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Dmitry Deniskin
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Anastasia Poliakova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Mikhail Chudinov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia; (L.V.); (K.K.); (A.L.); (M.Z.); (D.D.); (A.P.)
| | - Maxim Gureev
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg 194064, Russia;
| | - Marine Dogonadze
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, Saint Petersburg 191036, Russia; (M.D.); (T.V.)
| | - Tatiana Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, Saint Petersburg 191036, Russia; (M.D.); (T.V.)
| | - Elizaveta Rogacheva
- Pasteur Institute of Epidemiology and Microbiology, Saint Petersburg 197101, Russia; (E.R.); (L.K.)
| | - Lyudmila Kraeva
- Pasteur Institute of Epidemiology and Microbiology, Saint Petersburg 197101, Russia; (E.R.); (L.K.)
| | - Yuri Porozov
- Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, Moscow 125315, Russia;
- Advitam Laboratory, 11108 Belgrade, Serbia
| | - Viktor Korzhikov-Vlakh
- Department of Medical Chemistry, Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia
| |
Collapse
|
2
|
Ciaglia T, Miranda MR, Di Micco S, Vietri M, Smaldone G, Musella S, Di Sarno V, Auriemma G, Sardo C, Moltedo O, Pepe G, Bifulco G, Ostacolo C, Campiglia P, Manfra M, Vestuto V, Bertamino A. Neuroprotective Potential of Indole-Based Compounds: A Biochemical Study on Antioxidant Properties and Amyloid Disaggregation in Neuroblastoma Cells. Antioxidants (Basel) 2024; 13:1585. [PMID: 39765912 PMCID: PMC11673510 DOI: 10.3390/antiox13121585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Based on the established neuroprotective properties of indole-based compounds and their significant potential as multi-targeted therapeutic agents, a series of synthetic indole-phenolic compounds was evaluated as multifunctional neuroprotectors. Each compound demonstrated metal-chelating properties, particularly in sequestering copper ions, with quantitative analysis revealing approximately 40% chelating activity across all the compounds. In cellular models, these hybrid compounds exhibited strong antioxidant and cytoprotective effects, countering reactive oxygen species (ROS) generated by the Aβ(25-35) peptide and its oxidative byproduct, hydrogen peroxide, as demonstrated by quantitative analysis showing on average a 25% increase in cell viability and a reduction in ROS levels to basal states. Further analysis using thioflavin T fluorescence assays, circular dichroism, and computational studies indicated that the synthesized derivatives effectively promoted the self-disaggregation of the Aβ(25-35) fragment. Taken together, these findings suggest a unique profile of neuroprotective actions for indole-phenolic derivatives, combining chelating, antioxidant, and anti-aggregation properties, which position them as promising compounds for the development of multifunctional agents in Alzheimer's disease therapy. The methods used provide reliable in vitro data, although further in vivo validation and assessment of blood-brain barrier penetration are needed to confirm therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS), Via Salvatore de Renzi 50, 84125 Salerno, Italy;
| | - Mariapia Vietri
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carla Sardo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| |
Collapse
|
3
|
Sundriyal S. Basic nitrogen (BaN): a 'privileged element' in medicinal chemistry. Future Med Chem 2024; 16:2069-2071. [PMID: 39373180 PMCID: PMC11559362 DOI: 10.1080/17568919.2024.2409627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024] Open
Affiliation(s)
- Sandeep Sundriyal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, 333031, India
| |
Collapse
|
4
|
Espinosa-Bustos C, Bertrand J, Villegas-Menares A, Guerrero S, Di Marcotullio L, Navacci S, Schulte G, Kozielewicz P, Bloch N, Villela V, Paulino M, Kogan MJ, Cantero J, Salas CO. New Smoothened ligands based on the purine scaffold as potential agents for treating pancreatic cancer. Bioorg Chem 2024; 151:107681. [PMID: 39106711 DOI: 10.1016/j.bioorg.2024.107681] [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: 03/28/2024] [Revised: 06/27/2024] [Accepted: 07/27/2024] [Indexed: 08/09/2024]
Abstract
Aberrant activation of the Hedgehog (Hh) signalling pathway has been associated with the development and progression of pancreatic cancer. For this reason, blockade of Hh pathway by inhibitors targeting the G protein-coupled receptor Smoothened (SMO) has been considered as a therapeutic target for the treatment of this cancer. In our previous work, we obtained a new SMO ligand based on a purine scaffold (compound I), which showed interesting antitumor activity in several cancer cell lines. In this work, we report the design and synthesis of 17 new purine derivatives, some of which showed high cytotoxic effect on Mia-PaCa-2 (Hh-dependent pancreatic cancer cell lines) and low toxicity on non-neoplastic HEK-293 cells compared with gemcitabine, such as 8f, 8g and 8h (IC50 = 4.56, 4.11 and 3.08 μM, respectively). Two of these purines also showed their ability to bind to SMO through NanoBRET assays (pKi = 5.17 for 8f and 5.01 for 8h), with higher affinities to compound I (pKi = 1.51). In addition, docking studies provided insight the purine substitution pattern is related to the affinity on SMO. Finally, studies of Hh inhibition for selected purines, using a transcriptional functional assay based on luciferase activity in NIH3T3 Shh-Light II cells, demonstrated that 8g reduced GLI activity with a IC50 = 6.4 μM as well as diminished the expression of Hh target genes in two specific Hh-dependent cell models, Med1 cells and Ptch1-/- mouse embryonic fibroblasts. Therefore, our results provide a platform for the design of SMO ligands that could be potential selective cytotoxic agents for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Christian Espinosa-Bustos
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, 702843 Santiago, Chile
| | - Jeanluc Bertrand
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, 702843 Santiago, Chile
| | - Alondra Villegas-Menares
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, 702843 Santiago, Chile
| | - Simón Guerrero
- Facultad de Medicina, Universidad de Atacama, 153601 Copiapó, Chile
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, Faculty Pharmacy and Medicine, Sapienza University, 00161 Rome, Italy; Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, 00161 Rome, Italy
| | - Shirin Navacci
- Department of Molecular Medicine, Faculty Pharmacy and Medicine, Sapienza University, 00161 Rome, Italy
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institute, 17165 Solna, Stockholm, Sweden
| | - Pawel Kozielewicz
- Department of Physiology and Pharmacology, Karolinska Institute, 17165 Solna, Stockholm, Sweden
| | - Nicolas Bloch
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, 702843 Santiago, Chile
| | - Valentina Villela
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, 702843 Santiago, Chile
| | - Margot Paulino
- Departamento DETEMA, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Marcelo J Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, 8380492 Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Universidad de Chile, 8380492 Santiago, Chile
| | - Jorge Cantero
- Departamento DETEMA, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Cristian O Salas
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, 702843 Santiago, Chile.
| |
Collapse
|
5
|
Sousa RCN, Confessor VPP, Da Silva AKB, Almeida AR, Pinheiro FASD, Ferreira LS. Biomimetic Chemical Reactions with Natural Products Using Metalloporphyrins and Salen Complexes as Catalysts: A Brief Review. Chem Biodivers 2024; 21:e202400668. [PMID: 38763894 DOI: 10.1002/cbdv.202400668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
The cytochrome P450 is a superfamily of hemoproteins mainly present in the liver and are versatile biocatalysts. They participate in the primary metabolism and biosynthesis of various secondary metabolites. Chemical catalysts are utilized to replicate the activities of enzymes. Metalloporphyrins and Salen complexes can contribute to the products' characterization and elucidate biotransformation processes, which are investigated during pre-clinical trials. These catalysts also help discover biologically active compounds and get better yields of products of industrial interest. This review aims to investigate which natural product classes are being investigated by biomimetic chemical models and the functionalities applied in the use of these catalysts. A limited number of studies were observed, with terpenes and alkaloids being the most investigated natural product classes. The research also revealed that Metalloporphyrins are still the most popular in the studies, and the identity and yield of the products obtained depend on the reaction system conditions.
Collapse
Affiliation(s)
- Rita C N Sousa
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, 59078-900, Natal-RN, Brazil
| | - Vitor P P Confessor
- Pharmacy Department, Federal University of Rio Grande do Norte, 59012-570, Natal-RN, Brazil
| | - Antonio K B Da Silva
- Pharmacy Department, Federal University of Rio Grande do Norte, 59012-570, Natal-RN, Brazil
| | - Addison R Almeida
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, 59078-900, Natal-RN, Brazil
- Pharmacy Department, Federal University of Rio Grande do Norte, 59012-570, Natal-RN, Brazil
| | | | - Leandro S Ferreira
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, 59078-900, Natal-RN, Brazil
- Pharmacy Department, Federal University of Rio Grande do Norte, 59012-570, Natal-RN, Brazil
| |
Collapse
|
6
|
Sheng XH, Han LC, Gong A, Meng XS, Wang XH, Teng LS, Sun XH, Xu KC, Liu ZH, Wang T, Ma JP, Zhang L. Discovery of Novel Ortho-Aminophenol Derivatives Targeting Lipid Peroxidation with Potent Antiferroptotic Activities. J Med Chem 2024; 67:9536-9551. [PMID: 38822802 DOI: 10.1021/acs.jmedchem.4c00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
The concept of ferroptosis inhibition has gained growing recognition as a promising therapeutic strategy for addressing a wide range of diseases. Here, we present the discovery of four series of ortho-aminophenol derivatives as potential ferroptosis inhibitors beginning with the endogenous substance 3-hydroxyanthranilic acid (3-HA) by employing quantum chemistry techniques, in vitro and in vivo assays. Our findings reveal that these ortho-aminophenol derivatives exhibit unique intra-H bond interactions, compelling ortho-amines to achieve enhanced alignment with the aromatic π-system, thereby expanding their activity. Notably, compounds from all four series display remarkable activity against RSL3-induced ferroptosis, showcasing an activity 100 times more than that of 3-HA. Furthermore, these compounds also demonstrate robust in vivo efficacy in protecting mice from kidney ischemia-reperfusion injury and acetaminophen-induced hepatotoxicity. In summary, we provide four distinct series of active scaffolds that significantly expand the chemical space of ferroptosis inhibitors, serving as valuable insights for future structural modifications.
Collapse
Affiliation(s)
- Xie-Huang Sheng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Li-Cong Han
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Ao Gong
- Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Xiang-Shuai Meng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Xin-Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Lin-Song Teng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan 250014, China
| | - Xiao-Han Sun
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Kuo-Chen Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan 250014, China
| | - Zhao-Hua Liu
- The Model Animal Research Center, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Ting Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Jian-Ping Ma
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Lei Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan 250014, China
| |
Collapse
|
7
|
Chua HM, Moshawih S, Kifli N, Goh HP, Ming LC. Insights into the computer-aided drug design and discovery based on anthraquinone scaffold for cancer treatment: A systematic review. PLoS One 2024; 19:e0301396. [PMID: 38776291 PMCID: PMC11111074 DOI: 10.1371/journal.pone.0301396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND In the search for better anticancer drugs, computer-aided drug design (CADD) techniques play an indispensable role in facilitating the lengthy and costly drug discovery process especially when natural products are involved. Anthraquinone is one of the most widely-recognized natural products with anticancer properties. This review aimed to systematically assess and synthesize evidence on the utilization of CADD techniques centered on the anthraquinone scaffold for cancer treatment. METHODS The conduct and reporting of this review were done in accordance to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) 2020 guideline. The protocol was registered in the "International prospective register of systematic reviews" database (PROSPERO: CRD42023432904) and also published recently. The search strategy was designed based on the combination of concept 1 "CADD or virtual screening", concept 2 "anthraquinone" and concept 3 "cancer". The search was executed in PubMed, Scopus, Web of Science and MedRxiv on 30 June 2023. RESULTS Databases searching retrieved a total of 317 records. After deduplication and applying the eligibility criteria, the final review ended up with 32 articles in which 3 articles were found by citation searching. The CADD methods used in the studies were either structure-based alone (69%) or combined with ligand-based methods via parallel (9%) or sequential (22%) approaches. Molecular docking was performed in all studies, with Glide and AutoDock being the most popular commercial and public software used respectively. Protein data bank was used in most studies to retrieve the crystal structure of the targets of interest while the main ligand databases were PubChem and Zinc. The utilization of in-silico techniques has enabled a deeper dive into the structural, biological and pharmacological properties of anthraquinone derivatives, revealing their remarkable anticancer properties in an all-rounded fashion. CONCLUSION By harnessing the power of computational tools and leveraging the natural diversity of anthraquinone compounds, researchers can expedite the development of better drugs to address the unmet medical needs in cancer treatment by improving the treatment outcome for cancer patients.
Collapse
Affiliation(s)
- Hui Ming Chua
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Said Moshawih
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Nurolaini Kifli
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Hui Poh Goh
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
- School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
8
|
Tang SA, Fults A, Boyd SR, Gattu N, Tran KA, Fan J, MacKenzie KR, Palzkill T, Young DW, Chamakuri S. Expanding Complex Morpholines Using Systematic Chemical Diversity. Org Lett 2024; 26:3493-3497. [PMID: 38506470 DOI: 10.1021/acs.orglett.4c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
The morpholine heterocycle is a structural unit found in many bioactive compounds and FDA-approved drugs, but the generation of more complex C-functionalized morpholine derivatives remains considerably underexplored. Using systematic chemical diversity (SCD), a concept that guides the expansion of saturated drug-like scaffolds through regiochemical and stereochemical variation, we describe the synthesis of a collection of methyl-substituted morpholine acetic acid esters starting from enantiomerically pure amino acids and amino alcohols. In total, 24 diverse substituted morpholines were produced that vary systematically in regiochemistry and stereochemistry (relative and absolute). These diverse C-substituted morpholines can be directly applied in fragment screening or incorporated as building blocks in medicinal chemistry and library synthesis.
Collapse
Affiliation(s)
- Sunny Ann Tang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Afton Fults
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Shelton R Boyd
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Nikhil Gattu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Kevin A Tran
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Jiayi Fan
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Damian W Young
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Srinivas Chamakuri
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| |
Collapse
|
9
|
González JF, Dea-Ayuela MA, Huck L, Orduña JM, Bolás-Fernández F, de la Cuesta E, Haseen N, Mohammed AA, Menéndez JC. Dual Antitubercular and Antileishmanial Profiles of Quinoxaline Di- N-Oxides Containing an Amino Acidic Side Chain. Pharmaceuticals (Basel) 2024; 17:487. [PMID: 38675447 PMCID: PMC11054274 DOI: 10.3390/ph17040487] [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: 03/19/2024] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
We present a new category of quinoxaline di-N-oxides (QdNOs) containing amino acid side chains with dual antituberculosis and antileishmanial activity. These compounds were synthesized by combining a regioselective 2,5-piperazinedione opening and a Beirut reaction and were screened for their activity against Mycobacterium tuberculosis and the promastigote and amastigote forms of representative species of the Leishmania genus. Most QdNOs exhibited promising antitubercular activity with IC50 values ranging from 4.28 to 49.95 μM, comparable to clinically established drugs. Structure-activity relationship analysis emphasized the importance of substituents on the aromatic ring and the side chain. Antileishmanial tests showed that some selected compounds exhibited activity comparable to the positive control miltefosine against promastigotes of Leishmania amazonensis and Leishmania donovani. Notably, some compounds were found to be also more potent and less toxic than miltefosine in intracellular amastigote assays against Leishmania amazonensis. The compound showing the best dual antitubercular and leishmanicidal profile and a good selectivity index, 4h, can be regarded as a hit compound that opens up new opportunities for the development of integrated therapies against co-infections.
Collapse
Affiliation(s)
- Juan F. González
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain; (J.F.G.); (L.H.); (J.M.O.); (E.d.l.C.)
| | - María-Auxiliadora Dea-Ayuela
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, c/Santiago Ramón y Cajal, Alfara del Patriarca, 46115 Valencia, Spain;
| | - Lena Huck
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain; (J.F.G.); (L.H.); (J.M.O.); (E.d.l.C.)
| | - José María Orduña
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain; (J.F.G.); (L.H.); (J.M.O.); (E.d.l.C.)
| | - Francisco Bolás-Fernández
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain;
| | - Elena de la Cuesta
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain; (J.F.G.); (L.H.); (J.M.O.); (E.d.l.C.)
| | - Nazia Haseen
- AMIPRO SDN.BHD. Level 3, Bangunan Inkubator Universiti, Sains@USM, Lebuh Bukit Jambul, Bayan Lepas 11900, Pulau Pinang, Malaysia; (N.H.); (A.A.M.)
| | - Ashraf Ali Mohammed
- AMIPRO SDN.BHD. Level 3, Bangunan Inkubator Universiti, Sains@USM, Lebuh Bukit Jambul, Bayan Lepas 11900, Pulau Pinang, Malaysia; (N.H.); (A.A.M.)
| | - J. Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain; (J.F.G.); (L.H.); (J.M.O.); (E.d.l.C.)
| |
Collapse
|
10
|
Nitulescu GM, Stancov G, Seremet OC, Nitulescu G, Mihai DP, Duta-Bratu CG, Barbuceanu SF, Olaru OT. The Importance of the Pyrazole Scaffold in the Design of Protein Kinases Inhibitors as Targeted Anticancer Therapies. Molecules 2023; 28:5359. [PMID: 37513232 PMCID: PMC10385367 DOI: 10.3390/molecules28145359] [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: 06/18/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The altered activation or overexpression of protein kinases (PKs) is a major subject of research in oncology and their inhibition using small molecules, protein kinases inhibitors (PKI) is the best available option for the cure of cancer. The pyrazole ring is extensively employed in the field of medicinal chemistry and drug development strategies, playing a vital role as a fundamental framework in the structure of various PKIs. This scaffold holds major importance and is considered a privileged structure based on its synthetic accessibility, drug-like properties, and its versatile bioisosteric replacement function. It has proven to play a key role in many PKI, such as the inhibitors of Akt, Aurora kinases, MAPK, B-raf, JAK, Bcr-Abl, c-Met, PDGFR, FGFRT, and RET. Of the 74 small molecule PKI approved by the US FDA, 8 contain a pyrazole ring: Avapritinib, Asciminib, Crizotinib, Encorafenib, Erdafitinib, Pralsetinib, Pirtobrutinib, and Ruxolitinib. The focus of this review is on the importance of the unfused pyrazole ring within the clinically tested PKI and on the additional required elements of their chemical structures. Related important pyrazole fused scaffolds like indazole, pyrrolo[1,2-b]pyrazole, pyrazolo[4,3-b]pyridine, pyrazolo[1,5-a]pyrimidine, or pyrazolo[3,4-d]pyrimidine are beyond the subject of this work.
Collapse
Affiliation(s)
| | | | | | - Georgiana Nitulescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (G.M.N.)
| | | | | | | | | |
Collapse
|
11
|
Triethylammonium 2-(3-Hydroxy-2-oxoindolin-3-yl)-5,5-dimethyl-3-oxocyclohex-1-en-1-olate. MOLBANK 2023. [DOI: 10.3390/m1589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
In recent years, the application of privileged structures has become a powerful approach in the discovery of new biologically active molecules. Ion pairing is a strategy used to enhance the permeation of ionized topical drugs. A convenient and efficient method for the synthesis of triethylammonium 2-(3-hydroxy-2-oxoindolin-3-yl)-5,5-dimethyl-3-oxocyclohex-1-en-1-olate has been developed. The presented protocol includes an aldol reaction and the formation of an ammonium salt. Triethylamine is both a reactant and a catalyst in the process. The structure of the synthesized title compound has been established by 1H, 13C-NMR and IR spectroscopy, mass spectrometry, and elemental analysis.
Collapse
|
12
|
2,4-Diamino-5-(5-hydroxy-1-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-yl)-5H-chromeno[2,3-b]pyridine-3-carbonitrile. MOLBANK 2023. [DOI: 10.3390/m1541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
5H-Chromeno[2,3-b]pyridines are important compounds with industrial, biological, and medicinal properties. In this short note, the multicomponent reaction of salicylaldehyde, malononitrile dimer, and 2-phenyl-5-(trifluoromethyl)-2,4-dihydro-3H-pyrazol-3-one in dimethyl sulfoxide at ambient temperature was investigated to give 2,4-diamino-5-(5-hydroxy-1-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-yl)-5H-chromeno[2,3-b]pyridine-3-carbonitrile with good yield. The structure of the previously unknown chromeno[2,3-b]pyridine derivative was confirmed by elemental analysis, mass, nuclear magnetic resonance, and infrared spectroscopy data. The ADME (absorption, distribution, metabolism, and excretion) properties were also assessed.
Collapse
|
13
|
Hajjo R, Sabbah DA, Abusara OH, Al Bawab AQ. A Review of the Recent Advances in Alzheimer's Disease Research and the Utilization of Network Biology Approaches for Prioritizing Diagnostics and Therapeutics. Diagnostics (Basel) 2022; 12:diagnostics12122975. [PMID: 36552984 PMCID: PMC9777434 DOI: 10.3390/diagnostics12122975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a polygenic multifactorial neurodegenerative disease that, after decades of research and development, is still without a cure. There are some symptomatic treatments to manage the psychological symptoms but none of these drugs can halt disease progression. Additionally, over the last few years, many anti-AD drugs failed in late stages of clinical trials and many hypotheses surfaced to explain these failures, including the lack of clear understanding of disease pathways and processes. Recently, different epigenetic factors have been implicated in AD pathogenesis; thus, they could serve as promising AD diagnostic biomarkers. Additionally, network biology approaches have been suggested as effective tools to study AD on the systems level and discover multi-target-directed ligands as novel treatments for AD. Herein, we provide a comprehensive review on Alzheimer's disease pathophysiology to provide a better understanding of disease pathogenesis hypotheses and decipher the role of genetic and epigenetic factors in disease development and progression. We also provide an overview of disease biomarkers and drug targets and suggest network biology approaches as new tools for identifying novel biomarkers and drugs. We also posit that the application of machine learning and artificial intelligence to mining Alzheimer's disease multi-omics data will facilitate drug and biomarker discovery efforts and lead to effective individualized anti-Alzheimer treatments.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA
- National Center for Epidemics and Communicable Disease Control, Amman 11118, Jordan
- Correspondence:
| | - Dima A. Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Osama H. Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Abdel Qader Al Bawab
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| |
Collapse
|
14
|
Liu H, Lau VHM, Xu P, Chan TH, Huang Z. Diverse synthesis of α-tertiary amines and tertiary alcohols via desymmetric reduction of malonic esters. Nat Commun 2022; 13:4759. [PMID: 35963867 PMCID: PMC9376102 DOI: 10.1038/s41467-022-32560-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/03/2022] [Indexed: 11/27/2022] Open
Abstract
Amines and alcohols with a fully substituted α-carbon are structures of great value in organic synthesis and drug discovery. While conventional methods towards these motifs often rely on enantioselective carbon-carbon or carbon-heteroatom bond formation reactions, a desymmetric method is developed here by selectively hydrosilylating one of the esters of easily accessible α-substituted α-amino- and -oxymalonic esters. The desymmetrization is enabled by a suite of dinuclear zinc catalysts with pipecolinol-derived tetradentate ligands and can accommodate a diverse panel of heteroatom substituents, including secondary amides, tertiary amines, and ethers of different sizes. The polyfunctionalized reduction products, in return, have provided expeditious approaches to enantioenriched nitrogen- and oxygen-containing molecules, including dipeptides, vitamin analogs, and natural metabolites. Chiral α-tertiary amines and tertiary alcohols are prevalent in bioactive molecules yet challenging targets to access. Here, the authors provide a dinuclear zinc-catalyzed desymmetric approach based on readily available malonic esters.
Collapse
Affiliation(s)
- Haichao Liu
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Vincent Ho Man Lau
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Pan Xu
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Tsz Hin Chan
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Zhongxing Huang
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
15
|
Nitulescu GM. Quantitative and Qualitative Analysis of the Anti-Proliferative Potential of the Pyrazole Scaffold in the Design of Anticancer Agents. Molecules 2022; 27:molecules27103300. [PMID: 35630776 PMCID: PMC9146646 DOI: 10.3390/molecules27103300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
The current work presents an objective overview of the impact of one important heterocyclic structure, the pyrazole ring, in the development of anti-proliferative drugs. A set of 1551 pyrazole derivatives were extracted from the National Cancer Institute (NCI) database, together with their growth inhibition effects (GI%) on the NCI’s panel of 60 cancer cell lines. The structures of these derivatives were analyzed based on the compounds’ averages of GI% values across NCI-60 cell lines and the averages of the values for the outlier cells. The distribution and the architecture of the Bemis–Murcko skeletons were analyzed, highlighting the impact of certain scaffold structures on the anti-proliferative effect’s potency and selectivity. The drug-likeness, chemical reactivity and promiscuity risks of the compounds were predicted using AMDETlab. The pyrazole ring proved to be a versatile scaffold for the design of anticancer drugs if properly substituted and if connected with other cyclic structures. The 1,3-diphenyl-pyrazole emerged as a useful scaffold for potent and targeted anticancer candidates.
Collapse
Affiliation(s)
- George Mihai Nitulescu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania
| |
Collapse
|
16
|
Bian Y, Xie XQ. Artificial Intelligent Deep Learning Molecular Generative Modeling of Scaffold-Focused and Cannabinoid CB2 Target-Specific Small-Molecule Sublibraries. Cells 2022; 11:cells11050915. [PMID: 35269537 PMCID: PMC8909864 DOI: 10.3390/cells11050915] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 02/26/2022] [Indexed: 02/01/2023] Open
Abstract
Design and generation of high-quality target- and scaffold-specific small molecules is an important strategy for the discovery of unique and potent bioactive drug molecules. To achieve this goal, authors have developed the deep-learning molecule generation model (DeepMGM) and applied it for the de novo molecular generation of scaffold-focused small-molecule libraries. In this study, a recurrent neural network (RNN) using long short-term memory (LSTM) units was trained with drug-like molecules to result in a general model (g-DeepMGM). Sampling practices on indole and purine scaffolds illustrate the feasibility of creating scaffold-focused chemical libraries based on machine intelligence. Subsequently, a target-specific model (t-DeepMGM) for cannabinoid receptor 2 (CB2) was constructed following the transfer learning process of known CB2 ligands. Sampling outcomes can present similar properties to the reported active molecules. Finally, a discriminator was trained and attached to the DeepMGM to result in an in silico molecular design-test circle. Medicinal chemistry synthesis and biological validation was performed to further investigate the generation outcome, showing that XIE9137 was identified as a potential allosteric modulator of CB2. This study demonstrates how recent progress in deep learning intelligence can benefit drug discovery, especially in de novo molecular design and chemical library generation.
Collapse
Affiliation(s)
- Yuemin Bian
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence:
| |
Collapse
|
17
|
Choo MZY, Chai CLL. Promoting GAINs (Give Attention to Limitations in Assays) over PAINs Alerts: no PAINS, more GAINs. ChemMedChem 2022; 17:e202100710. [PMID: 35146933 DOI: 10.1002/cmdc.202100710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/14/2022] [Indexed: 11/09/2022]
Abstract
Many concepts and guidelines in medicinal chemistry have been introduced to aid in successful drug discovery and development. An example is the concept of Pan-Assay Interference Compounds (PAINS) and the elimination of such nuisance compounds from high-throughput screening (HTS) libraries. PAINs, along with other guidelines in medicinal chemistry, are like double-edged swords. If used appropriately, they may be beneficial for drug discovery and development. However, rigid and blind use of such concepts can hinder productivity. In this perspective, we introduce GAINS (give attention to limitations in assays) and highlight its relevance for successful drug discovery.
Collapse
Affiliation(s)
- Malcolm Z Y Choo
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, Singapore, 117543, Singapore
| | - Christina L L Chai
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, Singapore, 117543, Singapore
| |
Collapse
|
18
|
N-Acylated and N-Alkylated 2-Aminobenzothiazoles Are Novel Agents That Suppress the Generation of Prostaglandin E2. Biomolecules 2022; 12:biom12020267. [PMID: 35204768 PMCID: PMC8961538 DOI: 10.3390/biom12020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/23/2022] Open
Abstract
The quest for novel agents to regulate the generation of prostaglandin E2 (PGE2) is of high importance because this eicosanoid is a key player in inflammatory diseases. We synthesized a series of N-acylated and N-alkylated 2-aminobenzothiazoles and related heterocycles (benzoxazoles and benzimidazoles) and evaluated their ability to suppress the cytokine-stimulated generation of PGE2 in rat mesangial cells. 2-Aminobenzothiazoles, either acylated by the 3-(naphthalen-2-yl)propanoyl moiety (GK510) or N-alkylated by a chain carrying a naphthalene (GK543) or a phenyl moiety (GK562) at a distance of three carbon atoms, stand out in inhibiting PGE2 generation, with EC50 values ranging from 118 nM to 177 nM. Both GK510 and GK543 exhibit in vivo anti-inflammatory activity greater than that of indomethacin. Thus, N-acylated or N-alkylated 2-aminobenzothiazoles are novel leads for the regulation of PGE2 formation.
Collapse
|
19
|
Davies C, Shaaban S, Waldmann H. Asymmetric catalysis with chiral cyclopentadienyl complexes to access privileged scaffolds. TRENDS IN CHEMISTRY 2022. [DOI: 10.1016/j.trechm.2022.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
20
|
Li W, Zhang J, Wang M, Dong R, Zhou X, Zheng X, Sun L. Pyrimidine-fused Dinitrogenous Penta-heterocycles as a Privileged Scaffold for Anti-Cancer Drug Discovery. Curr Top Med Chem 2022; 22:284-304. [PMID: 35021973 DOI: 10.2174/1568026622666220111143949] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 11/22/2022]
Abstract
Pyrimidine-fused derivatives that are the inextricable part of DNA and RNA play a key role in the normal life cycle of cells. Pyrimidine-fused dinitrogenous penta-heterocycles including pyrazolopyrimidines and imidazopyrimidines is a special class of pyrimidine-fused compounds contributing to an important portion in anti-cancer drug discovery, which have been discovered as core structure for promising anti-cancer agents used in clinic or clinical evaluations. Pyrimidine-fused dinitrogenous penta-heterocycles have become one privileged scaffold for anti-cancer drug discovery. This review consists of the recent progress of pyrimidine-fused dinitrogenous penta-heterocycles as anti-cancer agents and their synthetic strategies. In addition, this review also summarizes some key structure-activity relationships (SARs) of pyrimidine-fused dinitrogenous penta-heterocycle derivatives as anti-cancer agents.
Collapse
Affiliation(s)
- Wen Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jinyang Zhang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Min Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ru Dong
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xin Zhou
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xin Zheng
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Liping Sun
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
21
|
Bayya C, Dokala A, Manda S. Novel 6, 7-disubstituted 7H-purine analogues as potential EGFR/HER2 dual kinase inhibitors overcome lapatinib resistance: Design, synthesis,in-vitroandin-vivoevaluation. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
22
|
Congenericity of Claimed Compounds in Patent Applications. Molecules 2021; 26:molecules26175253. [PMID: 34500686 PMCID: PMC8433967 DOI: 10.3390/molecules26175253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/04/2022] Open
Abstract
A method is presented to analyze quantitatively the degree of congenericity of claimed compounds in patent applications. The approach successfully differentiates patents exemplified with highly congeneric compounds of a structurally compact and well defined chemical series from patents containing a more diverse set of compounds around a more vaguely described patent claim. An application to 750 common patents available in SureChEMBL, SureChEMBLccs and ChEMBL is presented and the congenericity of patent compounds in those different sources discussed.
Collapse
|
23
|
Zárate AM, Espinosa-Bustos C, Guerrero S, Fierro A, Oyarzún-Ampuero F, Quest AFG, Di Marcotullio L, Loricchio E, Caimano M, Calcaterra A, González-Quiroz M, Aguirre A, Meléndez J, Salas CO. A New Smoothened Antagonist Bearing the Purine Scaffold Shows Antitumour Activity In Vitro and In Vivo. Int J Mol Sci 2021; 22:8372. [PMID: 34445078 PMCID: PMC8395040 DOI: 10.3390/ijms22168372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
The Smoothened (SMO) receptor is the most druggable target in the Hedgehog (HH) pathway for anticancer compounds. However, SMO antagonists such as vismodegib rapidly develop drug resistance. In this study, new SMO antagonists having the versatile purine ring as a scaffold were designed, synthesised, and biologically tested to provide an insight to their mechanism of action. Compound 4s was the most active and the best inhibitor of cell growth and selectively cytotoxic to cancer cells. 4s induced cell cycle arrest, apoptosis, a reduction in colony formation and downregulation of PTCH and GLI1 expression. BODIPY-cyclopamine displacement assays confirmed 4s is a SMO antagonist. In vivo, 4s strongly inhibited tumour relapse and metastasis of melanoma cells in mice. In vitro, 4s was more efficient than vismodegib to induce apoptosis in human cancer cells and that might be attributed to its dual ability to function as a SMO antagonist and apoptosis inducer.
Collapse
Affiliation(s)
- Ana María Zárate
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile; (A.M.Z.); (A.F.)
| | - Christian Espinosa-Bustos
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile;
| | - Simón Guerrero
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone 1007, Independencia, Santiago 8380492, Chile; (S.G.); (F.O.-A.); (A.F.G.Q.)
- Instituto de Investigación Interdisciplinar en Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad SEK (I3CBSEK), Fernando Manterola 0789, Providencia, Santiago 7520317, Chile
| | - Angélica Fierro
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile; (A.M.Z.); (A.F.)
| | - Felipe Oyarzún-Ampuero
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone 1007, Independencia, Santiago 8380492, Chile; (S.G.); (F.O.-A.); (A.F.G.Q.)
- Departamento de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Andrew F. G. Quest
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone 1007, Independencia, Santiago 8380492, Chile; (S.G.); (F.O.-A.); (A.F.G.Q.)
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Program of Cellular and Molecular Biology, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | - Lucia Di Marcotullio
- Laboratory Affiliated to Insituto Pasteur Italia, Fondazione Cenci Bognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Elena Loricchio
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Miriam Caimano
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Matías González-Quiroz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile;
| | - Adam Aguirre
- Laboratorio de Medicina Traslacional, Fundación Arturo López Pérez, Rancagua 878, Lower Fifth Floor, Providencia, Santiago 8320000, Chile;
| | - Jaime Meléndez
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile;
| | - Cristian O. Salas
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile; (A.M.Z.); (A.F.)
| |
Collapse
|
24
|
Lei X, Lampiri P, Patil P, Angeli G, Neochoritis CG, Dömling A. A multicomponent tetrazolo indole synthesis. Chem Commun (Camb) 2021; 57:6652-6655. [PMID: 34128009 PMCID: PMC8259579 DOI: 10.1039/d1cc02384e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The ubiquitous presence of the indole fragment in natural products and drugs asks for ever novel syntheses. We report an unprecedented mild, two-step synthesis of 2-tetrazolo substituted indoles based on the Ugi-tetrazole reaction combined with an acidic ring closure. A gram-scale synthesis, a bioactive compound and further transformations were performed. A short, diverse, and scalable Ugi synthesis towards the bioactive tetrazolo indoles.![]()
Collapse
Affiliation(s)
- Xiaofang Lei
- University of Crete, Department of Chemistry, Heraklion, Greece. and University of Groningen, Department of Pharmacy, Drug Design group, Groningen, The Netherlands.
| | | | - Pravin Patil
- University of Groningen, Department of Pharmacy, Drug Design group, Groningen, The Netherlands.
| | - Giasemi Angeli
- University of Crete, Department of Chemistry, Heraklion, Greece.
| | | | - Alexander Dömling
- University of Groningen, Department of Pharmacy, Drug Design group, Groningen, The Netherlands.
| |
Collapse
|
25
|
Zhang L, Zhang G, Xu S, Song Y. Recent advances of quinones as a privileged structure in drug discovery. Eur J Med Chem 2021; 223:113632. [PMID: 34153576 DOI: 10.1016/j.ejmech.2021.113632] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 01/08/2023]
Abstract
Privileged structures are conductive to discover novel bioactive substances because they can bind to multiple targets with high affinity. Quinones are considered to be a privileged structure and useful template for the design of new compounds with potential pharmacological activity. This article presents the recent developments (2014-2021 update) of quinones in the fields of antitumor, antibacterial, antifungal, antiviral, anti-Alzheimer's disease (AD) and antimalarial, mainly focusing on biological activities, structural modification and mechanism of action.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacy, Jinan Second People's Hospital, 250001, 148 Jingyi Road, Jinan, PR China
| | - Guiying Zhang
- Department of Pharmacy, Rizhao People's Hospital, 276800, 126 Tai'an Road, Rizhao, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, PR China
| | - Yuning Song
- Department of Clinical Pharmacy, Qilu Hospital of Shandong University, 250012, Jinan, PR China.
| |
Collapse
|
26
|
Pennington LD, Muegge I. Holistic drug design for multiparameter optimization in modern small molecule drug discovery. Bioorg Med Chem Lett 2021; 41:128003. [DOI: 10.1016/j.bmcl.2021.128003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 01/28/2023]
|
27
|
Brambilla E, Leoni S, Abbiati G, Pirovano V, Rossi E. Formal Aza‐Diels−Alder Reactions of Spiroindolenines with Electronrich Dienes. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Elisa Brambilla
- Dipartimento di Scienze Farmaceutiche – Sezione di Chimica Generale e Organica “A. Marchesini” Università degli Studi di Milano Via Venezian 21 20133 Milano Italy
| | - Sara Leoni
- Dipartimento di Scienze Farmaceutiche – Sezione di Chimica Generale e Organica “A. Marchesini” Università degli Studi di Milano Via Venezian 21 20133 Milano Italy
| | - Giorgio Abbiati
- Dipartimento di Scienze Farmaceutiche – Sezione di Chimica Generale e Organica “A. Marchesini” Università degli Studi di Milano Via Venezian 21 20133 Milano Italy
| | - Valentina Pirovano
- Dipartimento di Scienze Farmaceutiche – Sezione di Chimica Generale e Organica “A. Marchesini” Università degli Studi di Milano Via Venezian 21 20133 Milano Italy
| | - Elisabetta Rossi
- Dipartimento di Scienze Farmaceutiche – Sezione di Chimica Generale e Organica “A. Marchesini” Università degli Studi di Milano Via Venezian 21 20133 Milano Italy
| |
Collapse
|
28
|
Faheem, Karan Kumar B, Venkata Gowri Chandra Sekhar K, Chander S, Kunjiappan S, Murugesan S. 1,2,3,4-Tetrahydroisoquinoline (THIQ) as privileged scaffold for anticancer de novo drug design. Expert Opin Drug Discov 2021; 16:1119-1147. [PMID: 33908322 DOI: 10.1080/17460441.2021.1916464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Cancer is a dreadful disorder that is emerging as one of the leading causes of mortality across the globe. The complex tumor environment, supplemented with drawbacks of the existing drugs, has made it a global health concern. The Tetrahydroisoquinoline (THIQ) ring holds an important position in medicinal chemistry due to its wide range of pharmacological properties. Several THIQ based natural products have been previously explored for their antitumor properties, making it a vital scaffold for anticancer drug design.Areas covered: This review article addresses the potential of THIQ as anticancer agents. Various medicinal chemistry strategies employed for the design and development of THIQ analogs as inhibitors or modulators of relevant anticancer targets have been discussed in detail. Moreover, the common strategies employed for the synthesis of the core scaffold are also highlighted.Expert opinion: Evidently, THIQs have tremendous potential in anticancer drug design. Some of these analogs exhibited potent activity against various cancer molecular targets. However, there are some drawbacks, such as selectivity that need addressing. The synthetic ease for constructing the core scaffold complimented with its reactivity makes it ideal for further structure-activity relationship studies. For these reasons, THIQ is a privileged scaffold for the design and development of novel anticancer agents.
Collapse
Affiliation(s)
- Faheem
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | - Banoth Karan Kumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | | | - Subhash Chander
- Amity Institute of Phytomedicine and Phytochemistry, Amity University Uttar Pradesh, Noida, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| |
Collapse
|
29
|
Dadras A, Rezvanfar MA, Beheshti A, Naeimi SS, Siadati SA. An Urgent Industrial Scheme both for Total Synthesis, and for Pharmaceutical Analytical Analysis of Umifenovir as an Anti-Viral API for Treatment of COVID-19. Comb Chem High Throughput Screen 2021; 25:838-846. [PMID: 33538665 DOI: 10.2174/1386207324666210203175631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND This paper is prepared to reveal about an urgent industrial scheme for a fast and facile total synthesis of umifenovir (arbidol) (by one-pot stages) as an antiviral agent for treating 2019-nCoV virus via inhibiting its viral replication in the human cells. As COVID-19, takes thousands of lives all around the world, it seems that the medicinal resources would not be enough to supply billions of peoples, currently living on the planet earth. Thus, this pandemic and its subsequent impacts on the natural order of our life, would be one of the most important threats against the entire human race. Aims & Objective: Due to this, in this project, we have made attempts to find an operative approach for synthesizing this compound as an active pharmaceutical ingredient (API), which showed it could be effective in inhibiting the newly emerged coronavirus. RESULTS The designed scheme uses relatively cheap precursors, and contains one pot stages, instead of seven time consuming, and more costly, linear steps. Also, it is tried to use safe and cheap solvents like water, and ethanol, instead of toxic ones like methanol, and pyridine which could cause rejection of the API in the organic volatile impurities (OVI) test of pharmacopeia analysis, as well as increasing the concern of inflammability, explosive, and carcinogenic properties of those common solvents. CONCLUSION The most important pharmaceutical analytical methods containing OVI test (mainly ethanol (about 171 ppm) much lower than the limits, by gas chromatography-Flame Ionization Detector (GC-FID) instrument), Assay content (about 99.6% by potentiometric titration), and related purity analysis (by High performance liquid chromatography-Ultraviolet Detector (HPLC-UV)) (about 99.8%) were performed and described to give a more clear industrial scheme.
Collapse
Affiliation(s)
- Akbar Dadras
- R&D Department, Tofigh Daru Research and Engineering Company, TPICO, Tehran, . Iran
| | - Mohammad Amin Rezvanfar
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, . Iran
| | | | | | - Seyyed Amir Siadati
- R&D Department, Tofigh Daru Research and Engineering Company, TPICO, Tehran, . Iran
| |
Collapse
|
30
|
da Silva ET, de Andrade GF, Araújo ADS, Lourenço MCS, de Souza MVN. Antibacterial activity of new substituted 4-N-alkylated-2-trifluoromethyl-quinoline analogues against sensitive and resistant Mycobacterium tuberculosis strains. Eur J Pharm Sci 2021; 157:105596. [DOI: 10.1016/j.ejps.2020.105596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/17/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022]
|
31
|
Lai J, Li X, Wang Y, Yin S, Zhou J, Liu Z. AIScaffold: A Web-Based Tool for Scaffold Diversification Using Deep Learning. J Chem Inf Model 2020; 61:1-6. [PMID: 33356237 DOI: 10.1021/acs.jcim.0c00867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Molecular scaffolds are widely used in drug design. Many methods and tools have been developed to utilize the information in scaffolds. Scaffold diversification is frequently used by medicinal chemists in tasks such as lead compound optimization, but tools for scaffold diversification are still lacking. Here, we propose AIScaffold (https://iaidrug.stonewise.cn), a web-based tool for scaffold diversification using the deep generative model. This tool can perform large-scale (up to 500,000 molecules) diversification in several minutes and recommend the top 500 (top 0.1%) molecules. Features such as site-specific diversification are also supported. This tool can facilitate the scaffold diversification process for medicinal chemists, thereby accelerating drug design.
Collapse
Affiliation(s)
- Junyong Lai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191 Beijing, P. R. China
| | - Xiangbin Li
- Stonewise, No. 19 Zhongguancun Street, Haidian District, 100080 Beijing, P. R. China
| | - Yanxing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191 Beijing, P. R. China
| | - Shiqiu Yin
- Stonewise, No. 19 Zhongguancun Street, Haidian District, 100080 Beijing, P. R. China
| | - Jielong Zhou
- Stonewise, No. 19 Zhongguancun Street, Haidian District, 100080 Beijing, P. R. China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191 Beijing, P. R. China
| |
Collapse
|
32
|
Simoben CV, Qaseem A, Moumbock AFA, Telukunta KK, Günther S, Sippl W, Ntie‐Kang F. Pharmacoinformatic Investigation of Medicinal Plants from East Africa. Mol Inform 2020; 39:e2000163. [PMID: 32964659 PMCID: PMC7685152 DOI: 10.1002/minf.202000163] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022]
Abstract
Medicinal plants have widely been used in the traditional treatment of ailments and have been proven effective. Their contribution still holds an important place in modern drug discovery due to their chemical, and biological diversities. However, the poor documentation of traditional medicine, in developing African countries for instance, can lead to the loss of knowledge related to such practices. In this study, we present the Eastern Africa Natural Products Database (EANPDB) containing the structural and bioactivity information of 1870 unique molecules isolated from about 300 source species from the Eastern African region. This represents the largest collection of natural products (NPs) from this geographical region, covering literature data of the period from 1962 to 2019. The computed physicochemical properties and toxicity profiles of each compound have been included. A comparative analysis of some physico-chemical properties like molecular weight, H-bond donor/acceptor, logPo/w , etc. as well scaffold diversity analysis has been carried out with other published NP databases. EANPDB was combined with the previously published Northern African Natural Products Database (NANPDB), to form a merger African Natural Products Database (ANPDB), containing ∼6500 unique molecules isolated from about 1000 source species (freely available at http://african-compounds.org). As a case study, latrunculins A and B isolated from the sponge Negombata magnifica (Podospongiidae) with previously reported antitumour activities, were identified via substructure searching as molecules to be explored as putative binders of histone deacetylases (HDACs).
Collapse
Affiliation(s)
- Conrad V. Simoben
- Institute of PharmacyMartin-Luther University of Halle-WittenbergKurt-Mothes-Str. 306120Halle/SaaleGermany
| | - Ammar Qaseem
- Institute of Pharmaceutical Sciences, Research Group Pharmaceutical BioinformaticsAlbert-Ludwigs-University FreiburgHermann-Herder-Straße 979104FreiburgGermany
| | - Aurélien F. A. Moumbock
- Institute of Pharmaceutical Sciences, Research Group Pharmaceutical BioinformaticsAlbert-Ludwigs-University FreiburgHermann-Herder-Straße 979104FreiburgGermany
| | - Kiran K. Telukunta
- ELIXIR@PSB, VIB-UGent Center for Plant Systems BiologyTechnologiepark 719052GhentBelgium
| | - Stefan Günther
- Institute of Pharmaceutical Sciences, Research Group Pharmaceutical BioinformaticsAlbert-Ludwigs-University FreiburgHermann-Herder-Straße 979104FreiburgGermany
| | - Wolfgang Sippl
- Institute of PharmacyMartin-Luther University of Halle-WittenbergKurt-Mothes-Str. 306120Halle/SaaleGermany
| | - Fidele Ntie‐Kang
- Institute of PharmacyMartin-Luther University of Halle-WittenbergKurt-Mothes-Str. 306120Halle/SaaleGermany
- Department of Chemistry, Faculty of ScienceUniversity of BueaP.O. Box 63Buea CM00237Cameroon
- Institut für BotanikTechnische Universität DresdenZellescherWeg 20b01217DresdenGermany
| |
Collapse
|
33
|
Chini MG, Giordano A, Potenza M, Terracciano S, Fischer K, Vaccaro MC, Colarusso E, Bruno I, Riccio R, Koeberle A, Werz O, Bifulco G. Targeting mPGES-1 by a Combinatorial Approach: Identification of the Aminobenzothiazole Scaffold to Suppress PGE 2 Levels. ACS Med Chem Lett 2020; 11:783-789. [PMID: 32435385 DOI: 10.1021/acsmedchemlett.9b00618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
Microsomal prostaglandin E2 synthase-1 (mPGES-1), the terminal enzyme responsible for the production of inducible prostaglandin E2, has become an attractive target for the treatment of inflammation and cancer pathologies. Starting from an aminobenzothiazole scaffold, used as an unprecedented chemical core for mPGES-1 inhibition, a Combinatorial Virtual Screening campaign was conducted, using the X-ray crystal structure of human mPGES-1. Two combinatorial libraries (6 × 104) were obtained by decorating the aminobenzothiazole scaffold with all acyl chlorides and boronates available at the Merck database. The scientific multidisciplinary approach included virtual screening workflow, synthesis, and biological evaluation and led to the identification of three novel aminobenzothiazoles 1, 3, and 13 acting as mPGES-1 inhibitors. The three disclosed hits are able to inhibit mPGES-1 in a cell-free system (IC50 = 1.4 ± 0.2, 0.7 ± 0.1, and 1.7 ± 0.2 μM, respectively), and all are endowed with antitumoral properties against A549 human cancer cell lines at micromolar concentrations (28.5 ± 1.1, 18.1 ± 0.8, and 19.2 ± 1.3 μM, respectively).
Collapse
Affiliation(s)
- Maria G. Chini
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, Isernia, I-86090, Italy
| | - Assunta Giordano
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
- Institute of Biomolecular Chemistry (ICB), Consiglio Nazionale delle Ricerche (CNR), Via Campi Flegrei 34, I-80078, Pozzuoli, Napoli, Italy
| | - Marianna Potenza
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Stefania Terracciano
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Katrin Fischer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Maria C. Vaccaro
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Ester Colarusso
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Ines Bruno
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Raffaele Riccio
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
- Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| |
Collapse
|
34
|
Moraes CB, Witt G, Kuzikov M, Ellinger B, Calogeropoulou T, Prousis KC, Mangani S, Di Pisa F, Landi G, Iacono LD, Pozzi C, Freitas-Junior LH, Dos Santos Pascoalino B, Bertolacini CP, Behrens B, Keminer O, Leu J, Wolf M, Reinshagen J, Cordeiro-da-Silva A, Santarem N, Venturelli A, Wrigley S, Karunakaran D, Kebede B, Pöhner I, Müller W, Panecka-Hofman J, Wade RC, Fenske M, Clos J, Alunda JM, Corral MJ, Uliassi E, Bolognesi ML, Linciano P, Quotadamo A, Ferrari S, Santucci M, Borsari C, Costi MP, Gul S. Accelerating Drug Discovery Efforts for Trypanosomatidic Infections Using an Integrated Transnational Academic Drug Discovery Platform. SLAS DISCOVERY 2020; 24:346-361. [PMID: 30784368 PMCID: PMC6484532 DOI: 10.1177/2472555218823171] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
According to the World Health Organization, more than 1 billion people are at risk of or are affected by neglected tropical diseases. Examples of such diseases include trypanosomiasis, which causes sleeping sickness; leishmaniasis; and Chagas disease, all of which are prevalent in Africa, South America, and India. Our aim within the New Medicines for Trypanosomatidic Infections project was to use (1) synthetic and natural product libraries, (2) screening, and (3) a preclinical absorption, distribution, metabolism, and excretion-toxicity (ADME-Tox) profiling platform to identify compounds that can enter the trypanosomatidic drug discovery value chain. The synthetic compound libraries originated from multiple scaffolds with known antiparasitic activity and natural products from the Hypha Discovery MycoDiverse natural products library. Our focus was first to employ target-based screening to identify inhibitors of the protozoan Trypanosoma brucei pteridine reductase 1 ( TbPTR1) and second to use a Trypanosoma brucei phenotypic assay that made use of the T. brucei brucei parasite to identify compounds that inhibited cell growth and caused death. Some of the compounds underwent structure-activity relationship expansion and, when appropriate, were evaluated in a preclinical ADME-Tox assay panel. This preclinical platform has led to the identification of lead-like compounds as well as validated hits in the trypanosomatidic drug discovery value chain.
Collapse
Affiliation(s)
- Carolina B Moraes
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil.,2 Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo-SP, Brazil
| | - Gesa Witt
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Maria Kuzikov
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Bernhard Ellinger
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Theodora Calogeropoulou
- 4 National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Kyriakos C Prousis
- 4 National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Stefano Mangani
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Flavio Di Pisa
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giacomo Landi
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lucia Dello Iacono
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Cecilia Pozzi
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lucio H Freitas-Junior
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil.,2 Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo-SP, Brazil
| | - Bruno Dos Santos Pascoalino
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil
| | - Claudia P Bertolacini
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil
| | - Birte Behrens
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Oliver Keminer
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Jennifer Leu
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Markus Wolf
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Jeanette Reinshagen
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Anabela Cordeiro-da-Silva
- 6 Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, Porto, Portugal
| | - Nuno Santarem
- 6 Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, Porto, Portugal
| | | | | | | | | | - Ina Pöhner
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Wolfgang Müller
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Joanna Panecka-Hofman
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany.,11 Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Rebecca C Wade
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany.,12 Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany.,13 Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany
| | - Martina Fenske
- 14 Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Joachim Clos
- 15 Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Elisa Uliassi
- 17 Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Pasquale Linciano
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Quotadamo
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Ferrari
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Santucci
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Borsari
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Paola Costi
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sheraz Gul
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| |
Collapse
|
35
|
Lazou M, Tarushi A, Gritzapis P, Psomas G. Transition metal complexes with a novel guanine-based (E)-2-(2-(pyridin-2-ylmethylene)hydrazinyl)quinazolin-4(3H)-one: Synthesis, characterization, interaction with DNA and albumins and antioxidant activity. J Inorg Biochem 2020; 206:111019. [DOI: 10.1016/j.jinorgbio.2020.111019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 10/25/2022]
|
36
|
Xiao Q, Liu J, Nie JH, Kong LB, Lin J, Yan SJ. Silver-catalyzed cascade reactions of 3-cyanochromone with 1,1-enediamines: synthesis of highly functionalized 2-(pyridin-3-yl)-chromeno[2,3-d]pyrimidines. Org Chem Front 2020. [DOI: 10.1039/d0qo00388c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A novel protocol for the construction of 2-(pyridin-3-yl)-chromeno[2,3-d]pyrimidines from 3-cyanochromone with 1,1-enediamines via an unprecedented cascade reaction has been developed by simply refluxing the reactants under the catalysis of silver carbonate.
Collapse
Affiliation(s)
- Qiang Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource (Yunnan University)
- Ministry of Education and Yunnan Province
- School of Chemical Science and Technology
- Yunnan University
- Kunming
| | - Jin Liu
- Key Laboratory of Medicinal Chemistry for Natural Resource (Yunnan University)
- Ministry of Education and Yunnan Province
- School of Chemical Science and Technology
- Yunnan University
- Kunming
| | - Jia-Hui Nie
- Key Laboratory of Medicinal Chemistry for Natural Resource (Yunnan University)
- Ministry of Education and Yunnan Province
- School of Chemical Science and Technology
- Yunnan University
- Kunming
| | - Ling-Bin Kong
- Key Laboratory of Medicinal Chemistry for Natural Resource (Yunnan University)
- Ministry of Education and Yunnan Province
- School of Chemical Science and Technology
- Yunnan University
- Kunming
| | - Jun Lin
- Key Laboratory of Medicinal Chemistry for Natural Resource (Yunnan University)
- Ministry of Education and Yunnan Province
- School of Chemical Science and Technology
- Yunnan University
- Kunming
| | - Sheng-Jiao Yan
- Key Laboratory of Medicinal Chemistry for Natural Resource (Yunnan University)
- Ministry of Education and Yunnan Province
- School of Chemical Science and Technology
- Yunnan University
- Kunming
| |
Collapse
|
37
|
O. Salas C, Zarate AM, Kryštof V, Mella J, Faundez M, Brea J, Loza MI, Brito I, Hendrychová D, Jorda R, Cabrera AR, Tapia RA, Espinosa-Bustos C. Promising 2,6,9-Trisubstituted Purine Derivatives for Anticancer Compounds: Synthesis, 3D-QSAR, and Preliminary Biological Assays. Int J Mol Sci 2019; 21:ijms21010161. [PMID: 31881717 PMCID: PMC6981454 DOI: 10.3390/ijms21010161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
We designed, synthesized, and evaluated novel 2,6,9-trisubstituted purine derivatives for their prospective role as antitumor compounds. Using simple and efficient methodologies, 31 compounds were obtained. We tested these compounds in vitro to draw conclusions about their cell toxicity on seven cancer cells lines and one non-neoplastic cell line. Structural requirements for antitumor activity on two different cancer cell lines were analyzed with SAR and 3D-QSAR. The 3D-QSAR models showed that steric properties could better explain the cytotoxicity of compounds than electronic properties (70% and 30% of contribution, respectively). From this analysis, we concluded that an arylpiperazinyl system connected at position 6 of the purine ring is beneficial for cytotoxic activity, while the use of bulky systems at position C-2 of the purine is not favorable. Compound 7h was found to be an effective potential agent when compared with a currently marketed drug, cisplatin, in four out of the seven cancer cell lines tested. Compound 7h showed the highest potency, unprecedented selectivity, and complied with all the Lipinski rules. Finally, it was demonstrated that 7h induced apoptosis and caused cell cycle arrest at the S-phase on HL-60 cells. Our study suggests that substitution in the purine core by arylpiperidine moiety is essential to obtain derivatives with potential anticancer activity.
Collapse
Affiliation(s)
- Cristian O. Salas
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago de Chile 702843, Chile; (A.M.Z.); (R.A.T.)
- Correspondence: (C.O.S.); (C.E.-B.); Tel.: +56-22-354-4427 (C.O.S.); +56-22-354-4838 (C.E.-B.)
| | - Ana Maria Zarate
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago de Chile 702843, Chile; (A.M.Z.); (R.A.T.)
| | - Vladimir Kryštof
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany AS CR, Slechtitelu 27, 783 71 Olomouc, Czech Republic; (V.K.); (D.H.); (R.J.)
| | - Jaime Mella
- Instituto de Química y Bioquímica, Facultad de Ciencias, Universidad de Valparaíso, 2360102, Av. Gran Bretaña 1111, Playa Ancha, Valparaíso, Casilla 5030, Chile;
| | - Mario Faundez
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago de Chile 702843, Chile;
| | - Jose Brea
- Innopharma Screening Platform-BioFarma Research Group, Centre for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela 15706, Spain; (J.B.); (M.I.L.)
| | - María Isabel Loza
- Innopharma Screening Platform-BioFarma Research Group, Centre for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela 15706, Spain; (J.B.); (M.I.L.)
| | - Ivan Brito
- Departamento de Química, Facultad de Ciencias Básicas, Universidad de Antofagasta, Av. Angamos 601, Antofagasta 1240000, Chile;
| | - Denisa Hendrychová
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany AS CR, Slechtitelu 27, 783 71 Olomouc, Czech Republic; (V.K.); (D.H.); (R.J.)
| | - Radek Jorda
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany AS CR, Slechtitelu 27, 783 71 Olomouc, Czech Republic; (V.K.); (D.H.); (R.J.)
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hnevotinská 5, 77900 Olomouc, Czech Republic
| | - Alan R. Cabrera
- Departamento de Química Inorgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago de Chile 702843, Chile;
| | - Ricardo A. Tapia
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago de Chile 702843, Chile; (A.M.Z.); (R.A.T.)
| | - Christian Espinosa-Bustos
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago de Chile 702843, Chile;
- Correspondence: (C.O.S.); (C.E.-B.); Tel.: +56-22-354-4427 (C.O.S.); +56-22-354-4838 (C.E.-B.)
| |
Collapse
|
38
|
Grygorenko OO, Volochnyuk DM, Ryabukhin SV, Judd DB. The Symbiotic Relationship Between Drug Discovery and Organic Chemistry. Chemistry 2019; 26:1196-1237. [PMID: 31429510 DOI: 10.1002/chem.201903232] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/19/2019] [Indexed: 12/20/2022]
Abstract
All pharmaceutical products contain organic molecules; the source may be a natural product or a fully synthetic molecule, or a combination of both. Thus, it follows that organic chemistry underpins both existing and upcoming pharmaceutical products. The reverse relationship has also affected organic synthesis, changing its landscape towards increasingly complex targets. This Review article sets out to give a concise appraisal of this symbiotic relationship between organic chemistry and drug discovery, along with a discussion of the design concepts and highlighting key milestones along the journey. In particular, criteria for a high-quality compound library design enabling efficient virtual navigation of chemical space, as well as rise and fall of concepts for its synthetic exploration (such as combinatorial chemistry; diversity-, biology-, lead-, or fragment-oriented syntheses; and DNA-encoded libraries) are critically surveyed.
Collapse
Affiliation(s)
- Oleksandr O Grygorenko
- Enamine Ltd., Chervonotkatska Street 78, Kiev, 02094, Ukraine.,Taras Shevchenko National University of Kiev, Volodymyrska Street 60, Kiev, 01601, Ukraine
| | - Dmitriy M Volochnyuk
- Enamine Ltd., Chervonotkatska Street 78, Kiev, 02094, Ukraine.,Taras Shevchenko National University of Kiev, Volodymyrska Street 60, Kiev, 01601, Ukraine.,Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Murmanska Street 5, Kiev, 02660, Ukraine
| | - Sergey V Ryabukhin
- Enamine Ltd., Chervonotkatska Street 78, Kiev, 02094, Ukraine.,Taras Shevchenko National University of Kiev, Volodymyrska Street 60, Kiev, 01601, Ukraine
| | - Duncan B Judd
- Awridian Ltd., Stevenage Bioscience Catalyst, Gunnelswood Road, Stevenage, Herts, SG1 2FX, UK
| |
Collapse
|
39
|
Hassan AH, Yoo SY, Lee KW, Yoon YM, Ryu HW, Jeong Y, Shin JS, Kang SY, Kim SY, Lee HH, Park BY, Lee KT, Lee YS. Repurposing mosloflavone/5,6,7-trimethoxyflavone-resveratrol hybrids: Discovery of novel p38-α MAPK inhibitors as potent interceptors of macrophage-dependent production of proinflammatory mediators. Eur J Med Chem 2019; 180:253-267. [DOI: 10.1016/j.ejmech.2019.07.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
|
40
|
Gour J, Gatadi S, Pooladanda V, Ghouse SM, Malasala S, Madhavi YV, Godugu C, Nanduri S. Facile synthesis of 1,2,3-triazole-fused indolo- and pyrrolo[1,4]diazepines, DNA-binding and evaluation of their anticancer activity. Bioorg Chem 2019; 93:103306. [PMID: 31586710 DOI: 10.1016/j.bioorg.2019.103306] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/09/2022]
Abstract
A facile synthetic strategy has been developed for the generation of structurally diverse N-fused heterocycles. The formation of fused 1,2,3-triazole indolo and pyrrolodiazepines proceeds through an initial Knoevenagel condensation followed by intramolecular azide-alkyne cycloaddition reaction at room temperature without recourse to the traditional Cu(I)-catalyzed azide-alkyne cycloadditions. The synthesized compounds were evaluated for their in vitro anti-cancer activity against the NCI 60 cell line panel. Among the tested compounds, 3a and 3h were found to exhibit potent inhibitory activity against many of the cell lines. Cell cycle analysis indicated that the compounds inhibit the cell cycle at sub G1 phase. The DNA- nano drop method, viscosity experiment and docking studies suggested these compounds possess DNA binding affinity.
Collapse
Affiliation(s)
- Jitendra Gour
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Srikanth Gatadi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Venkatesh Pooladanda
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Shaik Mahammad Ghouse
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Satyaveni Malasala
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Y V Madhavi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Chandraiah Godugu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Srinivas Nanduri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
41
|
Dantas RF, Evangelista TCS, Neves BJ, Senger MR, Andrade CH, Ferreira SB, Silva-Junior FP. Dealing with frequent hitters in drug discovery: a multidisciplinary view on the issue of filtering compounds on biological screenings. Expert Opin Drug Discov 2019; 14:1269-1282. [DOI: 10.1080/17460441.2019.1654453] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Rafael Ferreira Dantas
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Tereza Cristina Santos Evangelista
- LaSOPB – Laboratório de Síntese Orgânica e Prospecção Biológica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Junior Neves
- LabChem – Laboratory of Cheminformatics, Centro Universitário de Anápolis, UniEVANGÉLICA, Anápolis, Brazil
| | - Mario Roberto Senger
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Carolina Horta Andrade
- LabMol – Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Sabrina Baptista Ferreira
- LaSOPB – Laboratório de Síntese Orgânica e Prospecção Biológica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Floriano Paes Silva-Junior
- LaBECFar – Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
González JF, Alcántara AR, Doadrio AL, Sánchez-Montero JM. Developments with multi-target drugs for Alzheimer's disease: an overview of the current discovery approaches. Expert Opin Drug Discov 2019; 14:879-891. [PMID: 31165654 DOI: 10.1080/17460441.2019.1623201] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Alzheimer's disease (AD), the most common type of dementia among older adults, is a chronic neurodegenerative pathology that causes a progressive loss of cognitive functioning with a decline of rational skills. It is well known that AD is multifactorial, so there are many different pharmacological targets that can be pursued. Areas covered: The authors highlight the strategic value of privileged scaffolds in a multi-target lead compound generation against AD, exploring the concept of multi-target design, with a special emphasis on hybrid compounds. Hence, the most promising building blocks for designing and synthesizing hybrid anti-AD drugs are shown, while also presenting the more advanced hybrid compounds. Expert opinion: The available therapeutic arsenal for AD, designed under the traditional paradigm of 'one-drug/one target/one-disease', is based on the inhibition of brain acetylcholinesterase (AChE) to increase acetylcholine (ACh) levels. However, this classical approach has not been sufficiently effective when used to treat any multifactor-depending pathology (cancer, diabetes or AD). The multi-target drug concept has been quickly adopted by medicinal chemists. The basic research developments reported in recent years are a solid foundation that will pave the way for the construction of future AD therapeutics.
Collapse
Affiliation(s)
- Juan F González
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Andrés R Alcántara
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Antonio L Doadrio
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Jose María Sánchez-Montero
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
43
|
Lampkin BJ, Monteiro C, Powers ET, Bouc PM, Kelly JW, VanVeller B. A designed protein binding-pocket to control excited-state intramolecular proton transfer fluorescence. Org Biomol Chem 2019; 17:1076-1080. [PMID: 30534794 PMCID: PMC6549506 DOI: 10.1039/c8ob02673d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Excited-state intramolecular proton transfer involves a photochemical isomerization and creates the opportunity for the emission of two distinct wavelengths of light from a single fluorophore. The selectivity between these two wavelengths of emission is dependent on the environment around the fluorophore and suggests the possibility for ratiometric monitoring of protein microenvironments. Unfortunately, nonspecific binding of ESIPT fluorophores does not often lead to dramatic changes in the ratio between the two wavelengths of emission. A protein binding pocket was designed to selectively discriminate between the two channels of emission available to an ESIPT fluorophore. This work is significant because it demonstrates that specific interactions between the protein and the fluorophore are essential to realize strong ratiometric differences between the two possible wavelengths of emission. The design strategies proposed here lead to an ESIPT fluorophore that can discern subtle differences in the interface between two proteins.
Collapse
Affiliation(s)
- Bryan J Lampkin
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, USA.
| | - Cecilia Monteiro
- Department of Chemistry, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Evan T Powers
- Department of Chemistry, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Paige M Bouc
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, USA.
| | - Jeffery W Kelly
- Department of Chemistry, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Brett VanVeller
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, USA.
| |
Collapse
|
44
|
Zhang X, Huang QF, Zou WL, Li QZ, Feng X, Jia ZQ, Liu Y, Li JL, Wang QW. Synthetic approach to skeletally diverse nitrogen heterocycles from dicyano-2-methylenebut-3-enoates. Org Chem Front 2019. [DOI: 10.1039/c9qo00509a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Divergent synthesis of three types of azaheterocycles, including vinylaziridines, pyrroline and 2-aminopyridines, has been achieved through [2 + 1], formal-[4 + 1] and [5 + 1] annulations.
Collapse
Affiliation(s)
- Xiang Zhang
- Chengdu Institute of Organic Chemistry
- Chinese Academy of Sciences
- Chengdu 610041
- China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
| | - Qing-Fei Huang
- Chengdu Institute of Organic Chemistry
- Chinese Academy of Sciences
- Chengdu 610041
- China
- University of Chinese Academy of Sciences
| | - Wen-Lin Zou
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- Chengdu 610052
- PR China
| | - Qing-Zhu Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- Chengdu 610052
- PR China
| | - Xin Feng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- Chengdu 610052
- PR China
| | - Zhi-Qiang Jia
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- Chengdu 610052
- PR China
| | - Yue Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- Chengdu 610052
- PR China
| | - Jun-Long Li
- Chengdu Institute of Organic Chemistry
- Chinese Academy of Sciences
- Chengdu 610041
- China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province
| | - Qi-Wei Wang
- Chengdu Institute of Organic Chemistry
- Chinese Academy of Sciences
- Chengdu 610041
- China
- University of Chinese Academy of Sciences
| |
Collapse
|
45
|
Němec V, Hylsová M, Maier L, Flegel J, Sievers S, Ziegler S, Schröder M, Berger B, Chaikuad A, Valčíková B, Uldrijan S, Drápela S, Souček K, Waldmann H, Knapp S, Paruch K. Furo[3,2‐b]pyridine: A Privileged Scaffold for Highly Selective Kinase Inhibitors and Effective Modulators of the Hedgehog Pathway. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201810312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Václav Němec
- Department of Chemistry, CZ-OpenscreenMasaryk University Kamenice 5 Brno 625 00 Czech Republic
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
| | - Michaela Hylsová
- Department of Chemistry, CZ-OpenscreenMasaryk University Kamenice 5 Brno 625 00 Czech Republic
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
| | - Lukáš Maier
- Department of Chemistry, CZ-OpenscreenMasaryk University Kamenice 5 Brno 625 00 Czech Republic
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
| | - Jana Flegel
- Max-Planck-Institute für Molekulare PhysiologieAbteilung Chemische Biologie Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Sonja Sievers
- Max-Planck-Institute für Molekulare PhysiologieAbteilung Chemische Biologie Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Slava Ziegler
- Max-Planck-Institute für Molekulare PhysiologieAbteilung Chemische Biologie Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Martin Schröder
- Institute for Pharmaceutical ChemistryStructural Genomics ConsortiumJohann Wolfgang Goethe-University Max-von-Laue-Strasse 15 60438 Frankfurt am Main Germany
| | - Benedict‐Tilman Berger
- Institute for Pharmaceutical ChemistryStructural Genomics ConsortiumJohann Wolfgang Goethe-University Max-von-Laue-Strasse 15 60438 Frankfurt am Main Germany
| | - Apirat Chaikuad
- Institute for Pharmaceutical ChemistryStructural Genomics ConsortiumJohann Wolfgang Goethe-University Max-von-Laue-Strasse 15 60438 Frankfurt am Main Germany
| | - Barbora Valčíková
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
- Department of BiologyFaculty of MedicineMasaryk University Kamenice 5 Brno 625 00 Czech Republic
| | - Stjepan Uldrijan
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
- Department of BiologyFaculty of MedicineMasaryk University Kamenice 5 Brno 625 00 Czech Republic
| | - Stanislav Drápela
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
- Department of CytokineticsInstitute of Biophysics CAS Královopolská 135 Brno 612 65 Czech Republic
| | - Karel Souček
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
- Department of CytokineticsInstitute of Biophysics CAS Královopolská 135 Brno 612 65 Czech Republic
| | - Herbert Waldmann
- Max-Planck-Institute für Molekulare PhysiologieAbteilung Chemische Biologie Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Stefan Knapp
- Institute for Pharmaceutical ChemistryStructural Genomics ConsortiumJohann Wolfgang Goethe-University Max-von-Laue-Strasse 15 60438 Frankfurt am Main Germany
| | - Kamil Paruch
- Department of Chemistry, CZ-OpenscreenMasaryk University Kamenice 5 Brno 625 00 Czech Republic
- International Clinical Research CentreSt. Anne's University Hospital Pekařská 53 Brno 656 91 Czech Republic
| |
Collapse
|
46
|
Němec V, Hylsová M, Maier L, Flegel J, Sievers S, Ziegler S, Schröder M, Berger BT, Chaikuad A, Valčíková B, Uldrijan S, Drápela S, Souček K, Waldmann H, Knapp S, Paruch K. Furo[3,2-b]pyridine: A Privileged Scaffold for Highly Selective Kinase Inhibitors and Effective Modulators of the Hedgehog Pathway. Angew Chem Int Ed Engl 2018; 58:1062-1066. [PMID: 30569600 DOI: 10.1002/anie.201810312] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/16/2018] [Indexed: 12/15/2022]
Abstract
Reported is the identification of the furo[3,2-b]pyridine core as a novel scaffold for potent and highly selective inhibitors of cdc-like kinases (CLKs) and efficient modulators of the Hedgehog signaling pathway. Initially, a diverse target compound set was prepared by synthetic sequences based on chemoselective metal-mediated couplings, including assembly of the furo[3,2-b]pyridine scaffold by copper-mediated oxidative cyclization. Optimization of the subseries containing 3,5-disubstituted furo[3,2-b]pyridines afforded potent, cell-active, and highly selective inhibitors of CLKs. Profiling of the kinase-inactive subset of 3,5,7-trisubstituted furo[3,2-b]pyridines revealed sub-micromolar modulators of the Hedgehog pathway.
Collapse
Affiliation(s)
- Václav Němec
- Department of Chemistry, CZ-Openscreen, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
| | - Michaela Hylsová
- Department of Chemistry, CZ-Openscreen, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
| | - Lukáš Maier
- Department of Chemistry, CZ-Openscreen, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
| | - Jana Flegel
- Max-Planck-Institute für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Sonja Sievers
- Max-Planck-Institute für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Slava Ziegler
- Max-Planck-Institute für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Martin Schröder
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, 60438, Frankfurt am Main, Germany
| | - Benedict-Tilman Berger
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, 60438, Frankfurt am Main, Germany
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, 60438, Frankfurt am Main, Germany
| | - Barbora Valčíková
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Stjepan Uldrijan
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Stanislav Drápela
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Královopolská 135, Brno, 612 65, Czech Republic
| | - Karel Souček
- International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Královopolská 135, Brno, 612 65, Czech Republic
| | - Herbert Waldmann
- Max-Planck-Institute für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 15, 60438, Frankfurt am Main, Germany
| | - Kamil Paruch
- Department of Chemistry, CZ-Openscreen, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Pekařská 53, Brno, 656 91, Czech Republic
| |
Collapse
|
47
|
Abstract
The concept of repurposing previously approved medications to the treatment of new indications by taking advantage of off-target effects has gained traction in recent years, particularly in areas of medicine that do not offer large profits to pharmaceutical firms. As infectious disease discovery research has declined among large pharmaceutical companies, the potential payoff of repurposing has become attractive. The concept of repurposing previously approved medications to the treatment of new indications by taking advantage of off-target effects has gained traction in recent years, particularly in areas of medicine that do not offer large profits to pharmaceutical firms. As infectious disease discovery research has declined among large pharmaceutical companies, the potential payoff of repurposing has become attractive. From these efforts, the triphenylethylene class of selective estrogen receptor modulators related to tamoxifen has shown activity against a wide range of medically important human pathogens, including bacteria, fungi, parasites, and viruses. Because it has activity against many pathogens affecting people in resource-limited areas of the world, TAM and related drugs may be particularly useful. Here, we review the in vitro, in vivo, and mechanistic studies of the anti-infective activity of tamoxifen, toremifene, clomiphene, and their analogs. We also discuss the pharmacologic properties of this privileged scaffold and its potential utility in treating infectious diseases.
Collapse
|
48
|
Balakin KV, Filosa R, Lavrenov SN, Mkrtchyan AS, Nawrozkij MB, Novakov IA. Arbidol: a quarter-century after. Past, present and future of the original Russian antiviral. RUSSIAN CHEMICAL REVIEWS 2018. [DOI: 10.1070/rcr4791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The present review is concerned with the synthesis and structure–activity relationship studies of Arbidol and its structural analogues. The latter are roughly divided into several unequal parts: indole- and benzofuran-based compounds, benzimidazole and imidazopyridine bioisosteres and ring-expanded quinoline derivatives. Much attention is focused on various types of antiviral activity of the above-mentioned Arbidol congeners, as well as of the parent compound itself. Features of Arbidol synthesis and metabolic changes are also discussed.
The bibliography includes 166 references.
Collapse
|
49
|
Gour J, Gatadi S, Nagarsenkar A, Babu BN, Madhavi YV, Nanduri S. Synthesis of Indolo[1,2-b
]isoquinoline Derivatives by Lewis Acid-Catalyzed Intramolecular Friedel-Crafts Alkylation Reaction. European J Org Chem 2018. [DOI: 10.1002/ejoc.201800162] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jitendra Gour
- National Institute of Pharmaceutical Education and Research (NIPER); 500 037 Hyderabad India
| | - Srikanth Gatadi
- National Institute of Pharmaceutical Education and Research (NIPER); 500 037 Hyderabad India
| | - Atulya Nagarsenkar
- National Institute of Pharmaceutical Education and Research (NIPER); 500 037 Hyderabad India
| | - Bathini Nagendra Babu
- National Institute of Pharmaceutical Education and Research (NIPER); 500 037 Hyderabad India
| | - Y. V. Madhavi
- National Institute of Pharmaceutical Education and Research (NIPER); 500 037 Hyderabad India
| | - Srinivas Nanduri
- National Institute of Pharmaceutical Education and Research (NIPER); 500 037 Hyderabad India
| |
Collapse
|
50
|
Yang SM, Wang CY, Lin CK, Karanam P, Reddy GM, Tsai YL, Lin W. Diversity-Oriented Synthesis of Furo[3,2-c
]coumarins and Benzofuranyl Chromenones through Chemoselective Acylation/Wittig Reaction. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201711524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Shu-Mei Yang
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| | - Chein-Yi Wang
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| | - Chun-Kai Lin
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| | - Praneeth Karanam
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| | - Ganapuram Madhusudhan Reddy
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| | - Yi-Ling Tsai
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| | - Wenwei Lin
- Department of Chemistry; National Taiwan Normal University; 88, Sec. 4, Tingchow Road Taipei 11677 Taiwan, R.O.C
| |
Collapse
|