1
|
Corfu AI, Santarem N, Luelmo S, Mazza G, Greco A, Altomare A, Ferrario G, Nasta G, Keminer O, Aldini G, Tamborini L, Basilico N, Parapini S, Gul S, Cordeiro-da-Silva A, Conti P, Borsari C. Discovery of 1,3,4-Oxadiazole Derivatives as Broad-Spectrum Antiparasitic Agents. ACS Infect Dis 2024; 10:2222-2238. [PMID: 38717116 DOI: 10.1021/acsinfecdis.4c00181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Vector-borne parasitic diseases (VBPDs) pose a significant threat to public health on a global scale. Collectively, Human African Trypanosomiasis (HAT), Leishmaniasis, and Malaria threaten millions of people, particularly in developing countries. Climate change might alter the transmission and spread of VBPDs, leading to a global burden of these diseases. Thus, novel agents are urgently needed to expand therapeutic options and limit the spread of drug-resistant parasites. Herein, we report the development of broad-spectrum antiparasitic agents by screening a known library of antileishmanial and antimalarial compounds toward Trypanosoma brucei (T. brucei) and identifying a 1,3,4-oxadiazole derivative (19) as anti-T. brucei hit with predicted blood-brain barrier permeability. Subsequently, extensive structure-activity-relationship studies around the lipophilic tail of 19 led to a potent antitrypanosomal and antimalarial compound (27), with moderate potency also toward Leishmania infantum (L. infantum) and Leishmania tropica. In addition, we discovered a pan-active antiparasitic molecule (24), showing low-micromolar IC50s toward T. brucei and Leishmania spp. promastigotes and amastigotes, and nanomolar IC50 against Plasmodium falciparum, together with high selectivity for the parasites over mammalian cells (THP-1). Early ADME-toxicity assays were used to assess the safety profile of the compounds. Overall, we characterized 24 and 27, bearing the 1,3,4-oxadiazole privileged scaffold, as broad-spectrum low-toxicity agents for the treatment of VBPDs. An alkyne-substituted chemical probe (30) was synthesized and will be utilized in proteomics experiments aimed at deconvoluting the mechanism of action in the T. brucei parasite.
Collapse
Affiliation(s)
- Alexandra Ioana Corfu
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nuno Santarem
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sara Luelmo
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Gaia Mazza
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Alessandro Greco
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulio Ferrario
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulia Nasta
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Lucia Tamborini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Silvia Parapini
- Department of Biomedical Sciences for Health, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Anabela Cordeiro-da-Silva
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Chiara Borsari
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| |
Collapse
|
2
|
Dodds AC, Puddu S, Sutherland A. Thioarylation of anilines using dual catalysis: two-step synthesis of phenothiazines. Org Biomol Chem 2022; 20:5602-5614. [PMID: 35796590 DOI: 10.1039/d2ob01082h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A two-step synthesis of phenothiazines has been developed using a dual-catalytic ortho-thioarylation reaction of anilines as the key step. Activation of N-(2-bromophenylthio)succinimide was achieved using the super Lewis acid, iron(III) triflimide and the Lewis base, diphenyl selenide, resulting in an accelerated and efficient ortho-thioarylation reaction of various protected aniline derivatives and less reactive, unprotected analogues. The thioarylated adducts were then cyclised to the desired phenothiazines using either an Ullmann-Goldberg or Buchwald-Hartwig coupling reaction. The dual catalytic thioarylation and copper(I)-catalysed cyclisation approach was used for the four-step synthesis of methopromazine, a neuroleptic agent with antipsychotic activity.
Collapse
Affiliation(s)
- Amy C Dodds
- School of Chemistry, The Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Sabrina Puddu
- School of Chemistry, The Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Andrew Sutherland
- School of Chemistry, The Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
3
|
Rui X, Wang C, Si D, Hui X, Li K, Wen H, Li W, Liu J. One-Pot Tandem Access to Phenothiazine Derivatives from Acetanilide and 2-Bromothiophenol via Rhodium-Catalyzed C-H Thiolation and Copper-Catalyzed C-N Amination. J Org Chem 2021; 86:6622-6632. [PMID: 33881319 DOI: 10.1021/acs.joc.1c00403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A one-pot and step economic reaction involving Rh(III)-catalyzed C-H thiolation and relay Cu(II)-catalyzed C-N amination of acetanilide and 2-bromothiophenol is reported here, with several valuable phenothiazine products obtained. This synthesis protocol proceeds from easily starting materials, demonstrating high atom economy, broad substrate scope, and good yield. Furthermore, the directing group can be easily eliminated, and chlorpromazine is provided in a large scale; thus this synthesis protocol could be utilized to construct phenothiazine scaffolds.
Collapse
Affiliation(s)
- Xiyan Rui
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chao Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dongjuan Si
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xuechao Hui
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Keting Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongmei Wen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
4
|
Design, synthesis and evaluation of cholinesterase hybrid inhibitors using a natural steroidal alkaloid as precursor. Bioorg Chem 2021; 111:104893. [PMID: 33882364 DOI: 10.1016/j.bioorg.2021.104893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 01/05/2023]
Abstract
To date, Alzheimer's disease is the most alarming neurodegenerative disorder worldwide. This illness is multifactorial in nature and cholinesterase inhibitors have been the ones used in clinical treatments. In this context, many of these drugs selectively inhibit the acetylcholinesterase enzyme interacting in both the active site and the peripheric anionic site. Besides, some agents have exhibited extensive benefits being able to co-inhibit butyrylcholinesterase. In this contribution, a strategy previously explored by numerous authors is reported; the synthesis of hybrid cholinesterase inhibitors. This strategy uses a molecule of recognized high inhibitory activity (tacrine) together with a steroidal alkaloid of natural origin using different connectors. The biological assays demonstrated the improvement in the inhibitory activity compared to the alkaloidal precursor, together with the reinforcement of the interactions in multiple sites of the enzymatic cavity. This strategy should be explored and exploited in this area. Docking and molecular dynamic studies were performed to explain enzyme-ligand interactions, assisting a structure-activity relationship analysis.
Collapse
|
5
|
Balewski Ł, Sączewski F, Bednarski PJ, Wolff L, Nadworska A, Gdaniec M, Kornicka A. Synthesis, Structure and Cytotoxicity Testing of Novel 7-(4,5-dihydro-1 H-imidazol-2-yl)-2-aryl-6,7-dihydro-2 H-imidazo[2,1- c][1,2,4]triazol-3(5 H)-Imine Derivatives. Molecules 2020; 25:E5924. [PMID: 33327611 PMCID: PMC7765142 DOI: 10.3390/molecules25245924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 11/24/2022] Open
Abstract
The appropriate 1-arylhydrazinecarbonitriles 1a-c are subjected to the reaction with 2-chloro-4,5-dihydro-1H-imidazole (2), yielding 7-(4,5-dihydro-1H-imidazol-2-yl)-2-aryl-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-imines 3a-c, which are subsequently converted into the corresponding amides 4a-e, 8a-c, sulfonamides 5a-n, 9, ureas 6a-I, and thioureas 7a-d. The structures of the newly prepared derivatives 3a-c, 4a-e, 5a-n, 6a-i, 7a-d, 8a-c, and 9 are confirmed by IR, NMR spectroscopic data, as well as single-crystal X-ray analyses of 5e and 8c. The in vitro cytotoxic potency of these compounds is determined on a panel of human cancer cell lines, and the relationships between structure and antitumor activity are discussed. The most active 4-chloro-N-(2-(4-chlorophenyl)-7-(4,5-dihydro-1H-imidazol-2-yl)-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-ylidene)benzamide (4e) and N-(7-(4,5-dihydro-1H-imidazol-2-yl)-2-(p-tolyl)-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-ylidene)-[1,1'-biphenyl]-4-sulfonamide (5l) inhibits the growth of the cervical cancer SISO and bladder cancer RT-112 cell lines with IC50 values in the range of 2.38-3.77 μM. Moreover, N-(7-(4,5-dihydro-1H-imidazol-2-yl)-2-phenyl-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-ylidene)-4-phenoxybenzenesulfonamide (5m) has the best selectivity towards the SISO cell line and induces apoptosis in this cell line.
Collapse
Affiliation(s)
- Łukasz Balewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| | - Franciszek Sączewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| | - Patrick J. Bednarski
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, F.-L. Jahn Strasse 17, D-17489 Greifswald, Germany; (P.J.B.); (L.W.)
| | - Lisa Wolff
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, F.-L. Jahn Strasse 17, D-17489 Greifswald, Germany; (P.J.B.); (L.W.)
| | - Anna Nadworska
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| | - Maria Gdaniec
- Faculty of Chemistry, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
| | - Anita Kornicka
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| |
Collapse
|
6
|
Di Martino RMC, Pruccoli L, Bisi A, Gobbi S, Rampa A, Martinez A, Pérez C, Martinez-Gonzalez L, Paglione M, Di Schiavi E, Seghetti F, Tarozzi A, Belluti F. Novel Curcumin-Diethyl Fumarate Hybrid as a Dualistic GSK-3β Inhibitor/Nrf2 Inducer for the Treatment of Parkinson's Disease. ACS Chem Neurosci 2020; 11:2728-2740. [PMID: 32663009 PMCID: PMC8009478 DOI: 10.1021/acschemneuro.0c00363] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
Common
copathogenic factors, including oxidative stress and neuroinflammation,
are found to play a vital role in the development of neurodegenerative
disorders, including Alzheimer’s disease (AD) and Parkinson’s
disease (PD). Nowadays, owing to the multifactorial character of the
diseases, no effective therapies are available, thus underlying the
need for new strategies. Overexpression of the enzyme GSK-3β
and downregulation of the Nrf2/ARE pathway are responsible for a decrease
in antioxidant defense effects. These pieces of evidence underline
the usefulness of dual GSK-3β inhibitors/Nrf2 inducers. In this
regard, to design a dual modulator, the structures of a curcumin-based
analogue, as GSK-3β inhibitor, and a diethyl fumarate fragment,
as Nrf2 inducer, were combined. Among the hybrids, 5 and 6 proved to effectively inhibit GSK-3β, while 4 and 5 showed a marked ability to activate Nrf2
together to increase the neuronal resistance to oxidative stress.
These last pieces of evidence translated into specific neuroprotective
effects of 4 and 5 against PD pathological
events including neurotoxicity elicited by α-synuclein aggregates
and 6-hydroxydopamine. Hybrid 5 also showed neuroprotective
effects in a C. elegans model of PD where the activation
of GSK-3β is intimately involved in Nrf2 regulation. In summary, 5 emerged as an interesting multitarget derivative, valuable
to be exploited in a multitarget PD perspective.
Collapse
Affiliation(s)
- Rita Maria Concetta Di Martino
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Letizia Pruccoli
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Ana Martinez
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Concepción Pérez
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | - Maria Paglione
- Department of Biology, Agriculture and Food Science, National Research Council (CNR), Institute of Biosciences and BioResources (IBBR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Elia Di Schiavi
- Department of Biology, Agriculture and Food Science, National Research Council (CNR), Institute of Biosciences and BioResources (IBBR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Francesca Seghetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
7
|
Seghetti F, Di Martino RMC, Catanzaro E, Bisi A, Gobbi S, Rampa A, Canonico B, Montanari M, Krysko DV, Papa S, Fimognari C, Belluti F. Curcumin-1,2,3-Triazole Conjugation for Targeting the Cancer Apoptosis Machinery. Molecules 2020; 25:E3066. [PMID: 32635622 PMCID: PMC7412087 DOI: 10.3390/molecules25133066] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
The burden of neoplastic diseases is widely recognized as a severe cause of mortality. The clinical inadequacy of most anticancer therapeutics urgently prompted intense drug discovery efforts toward the identification of new chemical entities endowed with a potent and safe antitumor profile. In this scenario, targeting cancer cells apoptosis machinery has emerged as a relevant strategy, useful for tackling the emergence of drug resistance. On this basis, a small library of naturally inspired hybrid molecules was obtained by combining, through a click chemistry approach, "privileged" synthons such as curcumin scaffold and 1,2,3-triazole building block. Compound 1, bearing a para-fluoro phenyl moiety, showed low-micromolar potency against T acute lymphoblastic leukemia cell growth. More in-depth biologic studies demonstrated, for this analog, cell death-inducing properties associated with its capability to simultaneously activate both the receptor and the mitochondrial apoptosis cascades. This peculiar behavior offers promises for achieving an expanded anticancer effect, namely intense cytotoxic response coupled with reduced predisposition of chemoresistance insurgence. Altogether, this study allowed the identification of compound 1 as a lead compound worth to be progressed as an anticancer drug candidate.
Collapse
Affiliation(s)
- Francesca Seghetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (F.S.); (R.M.C.D.M.); (A.B.); (S.G.); (A.R.)
| | - Rita Maria Concetta Di Martino
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (F.S.); (R.M.C.D.M.); (A.B.); (S.G.); (A.R.)
| | - Elena Catanzaro
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (F.S.); (R.M.C.D.M.); (A.B.); (S.G.); (A.R.)
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (F.S.); (R.M.C.D.M.); (A.B.); (S.G.); (A.R.)
| | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (F.S.); (R.M.C.D.M.); (A.B.); (S.G.); (A.R.)
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Ca’ Le Suore, 2, 61029 Urbino, Italy; (B.C.); (M.M.); (S.P.)
| | - Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Ca’ Le Suore, 2, 61029 Urbino, Italy; (B.C.); (M.M.); (S.P.)
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Ca’ Le Suore, 2, 61029 Urbino, Italy; (B.C.); (M.M.); (S.P.)
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (F.S.); (R.M.C.D.M.); (A.B.); (S.G.); (A.R.)
| |
Collapse
|
8
|
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
|
9
|
Borsari C, Jiménez-Antón MD, Eick J, Bifeld E, Torrado JJ, Olías-Molero AI, Corral MJ, Santarem N, Baptista C, Severi L, Gul S, Wolf M, Kuzikov M, Ellinger B, Reinshagen J, Witt G, Linciano P, Tait A, Costantino L, Luciani R, Tejera Nevado P, Zander-Dinse D, Franco CH, Ferrari S, Moraes CB, Cordeiro-da-Silva A, Ponterini G, Clos J, Alunda JM, Costi MP. Discovery of a benzothiophene-flavonol halting miltefosine and antimonial drug resistance in Leishmania parasites through the application of medicinal chemistry, screening and genomics. Eur J Med Chem 2019; 183:111676. [DOI: 10.1016/j.ejmech.2019.111676] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/01/2019] [Accepted: 09/02/2019] [Indexed: 01/24/2023]
|
10
|
Ivasiv V, Albertini C, Gonçalves AE, Rossi M, Bolognesi ML. Molecular Hybridization as a Tool for Designing Multitarget Drug Candidates for Complex Diseases. Curr Top Med Chem 2019; 19:1694-1711. [DOI: 10.2174/1568026619666190619115735] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022]
Abstract
Molecular hybridization is a well-exploited medicinal chemistry strategy that aims to combine
two molecules (or parts of them) in a new, single chemical entity. Recently, it has been recognized
as an effective approach to design ligands able to modulate multiple targets of interest. Hybrid compounds
can be obtained by linking (presence of a linker) or framework integration (merging or fusing)
strategies. Although very promising to combat the multifactorial nature of complex diseases, the development
of molecular hybrids faces the critical issues of selecting the right target combination and the
achievement of a balanced activity towards them, while maintaining drug-like-properties. In this review,
we present recent case histories from our own research group that demonstrate why and how molecular
hybridization can be carried out to address the challenges of multitarget drug discovery in two therapeutic
areas that are Alzheimer’s and parasitic diseases. Selected examples spanning from linker- to fragment-
based hybrids will allow to discuss issues and consequences relevant to drug design.
Collapse
Affiliation(s)
- Viktoriya Ivasiv
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum − University of Bologna, I-40126, Bologna, Italy
| | - Claudia Albertini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum − University of Bologna, I-40126, Bologna, Italy
| | - Ana E. Gonçalves
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum − University of Bologna, I-40126, Bologna, Italy
| | - Michele Rossi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum − University of Bologna, I-40126, Bologna, Italy
| | - Maria L. Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum − University of Bologna, I-40126, Bologna, Italy
| |
Collapse
|
11
|
Triazole derivatives and their antiplasmodial and antimalarial activities. Eur J Med Chem 2019; 166:206-223. [PMID: 30711831 DOI: 10.1016/j.ejmech.2019.01.047] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/19/2019] [Accepted: 01/19/2019] [Indexed: 01/23/2023]
Abstract
Malaria, caused by protozoan parasites of the genus Plasmodium especially by the most prevalent parasite Plasmodium falciparum, represents one of the most devastating and common infectious disease globally. Nearly half of the world population is under the risk of being infected, and more than 200 million new clinical cases with around half a million deaths occur annually. Drug therapy is the mainstay of antimalarial therapy, yet current drugs are threatened by the development of resistance, so it's imperative to develop new antimalarials with great potency against both drug-susceptible and drug-resistant malaria. Triazoles, bearing a five-membered heterocyclic ring with three nitrogen atoms, exhibit promising in vitro antiplasmodial and in vivo antimalarial activities. Moreover, several triazole-based drugs have already used in clinics for the treatment of various diseases, demonstrating the excellent pharmaceutical profiles. Therefore, triazole derivatives have the potential for clinical deployment in the control and eradication of malaria. This review covers the recent advances of triazole derivatives especially triazole hybrids as potential antimalarials. The structure-activity relationship is also discussed to provide an insight for rational designs of more efficient antimalarial candidates.
Collapse
|