1
|
Azevedo Teotônio Cavalcanti MD, Da Silva Menezes KJ, Oliveira Viana JD, Oliveira Rios ÉD, Corrêa de Farias AG, Weber KC, Nogueira F, Dos Santos Nascimento IJ, de Moura RO. Current trends to design antimalarial drugs targeting N-myristoyltransferase. Future Microbiol 2024:1-18. [PMID: 39440556 DOI: 10.1080/17460913.2024.2412397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
Malaria is a disease caused by Plasmodium spp., of which Plasmodium falciparum and Plasmodium vivax are the most prevalent. Unfortunately, traditional and some current treatment regimens face growing protozoan resistance. Thus, searching for and exploring new drugs and targets is necessary. One of these is N-myristoyltransferase (NMT). This enzyme is responsible for the myristoylation of several protein substrates in eukaryotic cells, including Plasmodium spp., thus enabling the assembly of protein complexes and stabilization of protein-membrane interactions. Given the importance of this target in developing new antiparasitic drugs, this review aims to explore the recent advances in the design of antimalarial drugs to target Plasmodium NMT.
Collapse
Affiliation(s)
- Misael de Azevedo Teotônio Cavalcanti
- Postgraduate Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina, Grande-PB, Brazil
- Drug Development & Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina, Grande, 58429-500, Brazil
| | - Karla Joane Da Silva Menezes
- Drug Development & Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina, Grande, 58429-500, Brazil
- Postgraduate Program of Drug Development & Technology Innovation, Federal University of Paraíba, João Pessoa, 58051-900, Brazil
| | - Jéssika De Oliveira Viana
- Postgraduate Program of Chemistry, Department of Chemistry, Federal University of Paraíba, João Pessoa, 58051-970, Brazil
| | | | - Arthur Gabriel Corrêa de Farias
- Drug Development & Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina, Grande, 58429-500, Brazil
| | - Karen Cacilda Weber
- Postgraduate Program of Chemistry, Department of Chemistry, Federal University of Paraíba, João Pessoa, 58051-970, Brazil
| | - Fatima Nogueira
- Universidade NOVA de Lisboa, UNL, Global Health & Tropical Medicine, GHTM, Associate Laboratory in Translation & Innovation Towards Global Health, LAREAL, Instituto de Higiene e Medicina Tropical, IHMT, Rua da Junqueira 100, 1349-008, Lisboa, Portugal
- LAQV-REQUIMTE, MolSyn, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisboa, Portugal
| | - Igor José Dos Santos Nascimento
- Postgraduate Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina, Grande-PB, Brazil
- Drug Development & Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina, Grande, 58429-500, Brazil
- Cesmac University Center, Pharmacy Department, Maceió, 57051-180, Brazil
| | - Ricardo Olimpio de Moura
- Postgraduate Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina, Grande-PB, Brazil
- Drug Development & Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina, Grande, 58429-500, Brazil
- Postgraduate Program of Drug Development & Technology Innovation, Federal University of Paraíba, João Pessoa, 58051-900, Brazil
| |
Collapse
|
2
|
Call DH, Adjei JA, Pilgrim R, Jeong JW, Willis EV, Zegarra RA, Tapia NL, Osterhaus M, Vance JA, Voyton CM, Call JA, Pizarro SS, Morris JC, Christensen KA. A multiplexed high throughput screening assay using flow cytometry identifies glycolytic molecular probes in bloodstream form Trypanosoma brucei. Int J Parasitol Drugs Drug Resist 2024; 26:100557. [PMID: 39163740 PMCID: PMC11381906 DOI: 10.1016/j.ijpddr.2024.100557] [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: 04/04/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024]
Abstract
Kinetoplastid organisms, including Trypanosoma brucei, are a significant health burden in many tropical and semitropical countries. Much of their metabolism is poorly understood. To better study kinetoplastid metabolism, chemical probes that inhibit kinetoplastid enzymes are needed. To discover chemical probes, we have developed a high-throughput flow cytometry screening assay that simultaneously measures multiple glycolysis-relevant metabolites in live T. brucei bloodstream form parasites. We transfected parasites with biosensors that measure glucose, ATP, or glycosomal pH. The glucose and ATP sensors were FRET biosensors, while the pH sensor was a GFP-based biosensor. The pH sensor exhibited a different fluorescent profile from the FRET sensors, allowing us to simultaneously measure pH and either glucose or ATP. Cell viability was measured in tandem with the biosensors using thiazole red. We pooled sensor cell lines, loaded them onto plates containing a compound library, and then analyzed them by flow cytometry. The library was analyzed twice, once with the pooled pH and glucose sensor cell lines and once with the pH and ATP sensor cell lines. Multiplexing sensors provided some internal validation of active compounds and gave potential clues for each compound's target(s). We demonstrated this using the glycolytic inhibitor 2-deoxyglucose and the alternative oxidase inhibitor salicylhydroxamic acid. Individual biosensor-based assays exhibited a Z'-factor value acceptable for high-throughput screening, including when multiplexed. We tested assay performance in a pilot screen of 14,976 compounds from the Life Chemicals Compound Library. We obtained hit rates from 0.2 to 0.4% depending on the biosensor, with many compounds impacting multiple sensors. We rescreened 44 hits, and 28 (64%) showed repeatable activity for one or more sensors. One compound exhibited EC50 values in the low micromolar range against two sensors. We expect this method will enable the discovery of glycolytic chemical probes to improve metabolic studies in kinetoplastid parasites.
Collapse
Affiliation(s)
- Daniel H Call
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - John Asafo Adjei
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Ryan Pilgrim
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - James W Jeong
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - E Vance Willis
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Ronald A Zegarra
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Nicholas L Tapia
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Madalyn Osterhaus
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Jacob A Vance
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Charles M Voyton
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA; Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA.
| | - James A Call
- Chemistry and Biochemistry Department, Brigham Young University, Provo, UT, USA.
| | - Sabrina S Pizarro
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA; Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, USA.
| | - James C Morris
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA; Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, SC, USA.
| | | |
Collapse
|
3
|
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
|
4
|
Hudu SA, Jimoh AO, Adeshina KA, Otalike EG, Tahir A, Hegazy AA. An insight into the Success, Challenges, and Future perspectives of eliminating Neglected tropical disease. SCIENTIFIC AFRICAN 2024; 24:e02165. [DOI: 10.1016/j.sciaf.2024.e02165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2024] Open
|
5
|
Marín M, López M, Gallego-Yerga L, Álvarez R, Peláez R. Experimental structure based drug design (SBDD) applications for anti-leishmanial drugs: A paradigm shift? Med Res Rev 2024; 44:1055-1120. [PMID: 38142308 DOI: 10.1002/med.22005] [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: 05/04/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/25/2023]
Abstract
Leishmaniasis is a group of neglected tropical diseases caused by at least 20 species of Leishmania protozoa, which are spread by the bite of infected sandflies. There are three main forms of the disease: cutaneous leishmaniasis (CL, the most common), visceral leishmaniasis (VL, also known as kala-azar, the most serious), and mucocutaneous leishmaniasis. One billion people live in areas endemic to leishmaniasis, with an annual estimation of 30,000 new cases of VL and more than 1 million of CL. New treatments for leishmaniasis are an urgent need, as the existing ones are inefficient, toxic, and/or expensive. We have revised the experimental structure-based drug design (SBDD) efforts applied to the discovery of new drugs against leishmaniasis. We have grouped the explored targets according to the metabolic pathways they belong to, and the key achieved advances are highlighted and evaluated. In most cases, SBDD studies follow high-throughput screening campaigns and are secondary to pharmacokinetic optimization, due to the majoritarian belief that there are few validated targets for SBDD in leishmaniasis. However, some SBDD strategies have significantly contributed to new drug candidates against leishmaniasis and a bigger number holds promise for future development.
Collapse
Affiliation(s)
- Miguel Marín
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Marta López
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
6
|
Gonçalves RCR, Teixeira F, Peñalver P, Costa SPG, Morales JC, Raposo MMM. Designing Antitrypanosomal and Antileishmanial BODIPY Derivatives: A Computational and In Vitro Assessment. Molecules 2024; 29:2072. [PMID: 38731562 PMCID: PMC11085077 DOI: 10.3390/molecules29092072] [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: 03/13/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Leishmaniasis and Human African trypanosomiasis pose significant public health threats in resource-limited regions, accentuated by the drawbacks of the current antiprotozoal treatments and the lack of approved vaccines. Considering the demand for novel therapeutic drugs, a series of BODIPY derivatives with several functionalizations at the meso, 2 and/or 6 positions of the core were synthesized and characterized. The in vitro activity against Trypanosoma brucei and Leishmania major parasites was carried out alongside a human healthy cell line (MRC-5) to establish selectivity indices (SIs). Notably, the meso-substituted BODIPY, with 1-dimethylaminonaphthalene (1b) and anthracene moiety (1c), were the most active against L. major, displaying IC50 = 4.84 and 5.41 μM, with a 16 and 18-fold selectivity over MRC-5 cells, respectively. In contrast, the mono-formylated analogues 2b and 2c exhibited the highest toxicity (IC50 = 2.84 and 6.17 μM, respectively) and selectivity (SI = 24 and 11, respectively) against T. brucei. Further insights on the activity of these compounds were gathered from molecular docking studies. The results suggest that these BODIPYs act as competitive inhibitors targeting the NADPH/NADP+ linkage site of the pteridine reductase (PR) enzyme. Additionally, these findings unveil a range of quasi-degenerate binding complexes formed between the PRs and the investigated BODIPY derivatives. These results suggest a potential correlation between the anti-parasitic activity and the presence of multiple configurations that block the same site of the enzyme.
Collapse
Affiliation(s)
- Raquel C R Gonçalves
- Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Advanced (Magnetic) Theranostic Nanostructures Lab, International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Filipe Teixeira
- Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Pablo Peñalver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avenida del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - Susana P G Costa
- Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Juan C Morales
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avenida del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - M Manuela M Raposo
- Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
7
|
Machado I, Gambino D. Metallomics: An Essential Tool for the Study of Potential Antiparasitic Metallodrugs. ACS OMEGA 2024; 9:15744-15752. [PMID: 38617611 PMCID: PMC11007724 DOI: 10.1021/acsomega.3c10200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 04/16/2024]
Abstract
Metallomics is an emerging area of omics approaches that has grown enormously in the past few years. It integrates research related to metals in biological systems, in symbiosis with genomics and proteomics. These omics approaches can provide in-depth insights into the mechanisms of action of potential metallodrugs, including their physiological metabolism and their molecular targets. Herein, we review the most significant advances concerning cellular uptake and subcellular distribution assays of different potential metallodrugs with activity against Trypanosma cruzi, the protozoan parasite that causes Chagas disease, a pressing health problem in high-poverty areas of Latin America. Furthermore, the first multiomics approaches including metallomics, proteomics, and transcriptomics for the comprehensive study of potential metallodrugs with anti-Trypanosoma cruzi activity are described.
Collapse
Affiliation(s)
- Ignacio Machado
- Área
Química Analítica, Facultad de Química, Área Química
Inorgánica, Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay
| | - Dinorah Gambino
- Área
Química Analítica, Facultad de Química, Área Química
Inorgánica, Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay
| |
Collapse
|
8
|
Hamid A, Mäser P, Mahmoud AB. Drug Repurposing in the Chemotherapy of Infectious Diseases. Molecules 2024; 29:635. [PMID: 38338378 PMCID: PMC10856722 DOI: 10.3390/molecules29030635] [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: 12/18/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Repurposing is a universal mechanism for innovation, from the evolution of feathers to the invention of Velcro tape. Repurposing is particularly attractive for drug development, given that it costs more than a billion dollars and takes longer than ten years to make a new drug from scratch. The COVID-19 pandemic has triggered a large number of drug repurposing activities. At the same time, it has highlighted potential pitfalls, in particular when concessions are made to the target product profile. Here, we discuss the pros and cons of drug repurposing for infectious diseases and analyze different ways of repurposing. We distinguish between opportunistic and rational approaches, i.e., just saving time and money by screening compounds that are already approved versus repurposing based on a particular target that is common to different pathogens. The latter can be further distinguished into divergent and convergent: points of attack that are divergent share common ancestry (e.g., prokaryotic targets in the apicoplast of malaria parasites), whereas those that are convergent arise from a shared lifestyle (e.g., the susceptibility of bacteria, parasites, and tumor cells to antifolates due to their high rate of DNA synthesis). We illustrate how such different scenarios can be capitalized on by using examples of drugs that have been repurposed to, from, or within the field of anti-infective chemotherapy.
Collapse
Affiliation(s)
- Amal Hamid
- Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, 4123 Basel, Switzerland
- Faculty of Science, University of Basel, 4001 Basel, Switzerland
| | - Abdelhalim Babiker Mahmoud
- Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, 66123 Saarbruecken, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| |
Collapse
|
9
|
Jia H, Hu L, Zhang J, Huang X, Jiang Y, Dong G, Liu C, Liu X, Kim M, Zhan P. Recent advances of phenotypic screening strategies in the application of anti-influenza virus drug discovery. RSC Med Chem 2024; 15:70-80. [PMID: 38283223 PMCID: PMC10809416 DOI: 10.1039/d3md00513e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/07/2023] [Indexed: 01/30/2024] Open
Abstract
Seasonal and pandemic influenza virus infections not only pose a serious threat to human health but also cause tremendous economic losses and social burdens. However, due to the inherent high variability of influenza virus RNA genomes, the existing anti-influenza virus drugs have been frequently faced with the clinical issue of emerging drug-resistant mutants. Therefore, there is an urgent need to develop efficient and broad-spectrum antiviral agents against wild-type and drug-resistant mutant strains. Phenotypic screening has been widely employed as a reliable strategy to evaluate antiviral efficacy of novel agents independent of their modes of action, either directly targeting viral proteins or regulating cellular factors involved in the virus life cycle. Here, from the point of view of medicinal chemistry, we review the research progress of phenotypic screening strategies by focusing direct acting antivirals against influenza virus. It could provide scientific insights into discovery of a distinctive class of therapeutic candidates that ensure high efficiency but low cytotoxicity, and address issues from circulation of drug-resistant influenza viruses in the future.
Collapse
Affiliation(s)
- Huinan Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Lide Hu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Xing Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Yuanmin Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Guanyu Dong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
- Suzhou Research Institute of Shandong University Room 607, Building B of NUSP, No. 388 Ruoshui Road, SIP Suzhou Jiangsu 215123 P.R. China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT) Daejeon 34114 Korea
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University 44 West Culture Road 250012 Jinan Shandong P.R. China
| |
Collapse
|
10
|
Morales-Luna L, Vázquez-Bautista M, Martínez-Rosas V, Rojas-Alarcón MA, Ortega-Cuellar D, González-Valdez A, Pérez de la Cruz V, Arreguin-Espinosa R, Rodríguez-Bustamante E, Rodríguez-Flores E, Hernández-Ochoa B, Gómez-Manzo S. Fused Enzyme Glucose-6-Phosphate Dehydrogenase::6-Phosphogluconolactonase (G6PD::6PGL) as a Potential Drug Target in Giardia lamblia, Trichomonas vaginalis, and Plasmodium falciparum. Microorganisms 2024; 12:112. [PMID: 38257939 PMCID: PMC10819308 DOI: 10.3390/microorganisms12010112] [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: 12/04/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Several microaerophilic parasites such as Giardia lamblia, Trichomonas vaginalis, and Plasmodium falciparum are major disease-causing organisms and are responsible for spreading infections worldwide. Despite significant progress made in understanding the metabolism and molecular biology of microaerophilic parasites, chemotherapeutic treatment to control it has seen limited progress. A current proposed strategy for drug discovery against parasitic diseases is the identification of essential key enzymes of metabolic pathways associated with the parasite's survival. In these organisms, glucose-6-phosphate dehydrogenase::6-phosphogluconolactonase (G6PD:: 6PGL), the first enzyme of the pentose phosphate pathway (PPP), is essential for its metabolism. Since G6PD:: 6PGL provides substrates for nucleotides synthesis and NADPH as a source of reducing equivalents, it could be considered an anti-parasite drug target. This review analyzes the anaerobic energy metabolism of G. lamblia, T. vaginalis, and P. falciparum, with a focus on glucose metabolism through the pentose phosphate pathway and the significance of the fused G6PD:: 6PGL enzyme as a therapeutic target in the search for new drugs.
Collapse
Affiliation(s)
- Laura Morales-Luna
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Montserrat Vázquez-Bautista
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Víctor Martínez-Rosas
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Miriam Abigail Rojas-Alarcón
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Daniel Ortega-Cuellar
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Abigail González-Valdez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Verónica Pérez de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| | - Roberto Arreguin-Espinosa
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.A.-E.); (E.R.-B.); (E.R.-F.)
| | - Eduardo Rodríguez-Bustamante
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.A.-E.); (E.R.-B.); (E.R.-F.)
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Eden Rodríguez-Flores
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.A.-E.); (E.R.-B.); (E.R.-F.)
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City 06720, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
| |
Collapse
|
11
|
Bailey BL, Nguyen W, Cowman AF, Sleebs BE. Chemo-proteomics in antimalarial target identification and engagement. Med Res Rev 2023; 43:2303-2351. [PMID: 37232495 PMCID: PMC10947479 DOI: 10.1002/med.21975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 04/24/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
Humans have lived in tenuous battle with malaria over millennia. Today, while much of the world is free of the disease, areas of South America, Asia, and Africa still wage this war with substantial impacts on their social and economic development. The threat of widespread resistance to all currently available antimalarial therapies continues to raise concern. Therefore, it is imperative that novel antimalarial chemotypes be developed to populate the pipeline going forward. Phenotypic screening has been responsible for the majority of the new chemotypes emerging in the past few decades. However, this can result in limited information on the molecular target of these compounds which may serve as an unknown variable complicating their progression into clinical development. Target identification and validation is a process that incorporates techniques from a range of different disciplines. Chemical biology and more specifically chemo-proteomics have been heavily utilized for this purpose. This review provides an in-depth summary of the application of chemo-proteomics in antimalarial development. Here we focus particularly on the methodology, practicalities, merits, and limitations of designing these experiments. Together this provides learnings on the future use of chemo-proteomics in antimalarial development.
Collapse
Affiliation(s)
- Brodie L. Bailey
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - William Nguyen
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Alan F. Cowman
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Brad E. Sleebs
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
12
|
Man Chin C, Dos Santos JL. Special Issue "Drug Candidates for the Treatment of Infectious Diseases". Pharmaceuticals (Basel) 2023; 16:1257. [PMID: 37765065 PMCID: PMC10534658 DOI: 10.3390/ph16091257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Infectious diseases encompass a range of conditions stemming from parasites [...].
Collapse
Affiliation(s)
- Chung Man Chin
- School of Pharmaceutical Science, State University of São Paulo (Unesp), Araraquara 14800-903, Brazil
- Union of the Colleges of the Great Lakes (UNILAGO), School of Medicine, São José do Rio Preto 15030-070, Brazil
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Science, State University of São Paulo (Unesp), Araraquara 14800-903, Brazil
| |
Collapse
|
13
|
Pathak S, Bhardwaj M, Agrawal N, Bhardwaj A. A comprehensive review on potential candidates for the treatment of chagas disease. Chem Biol Drug Des 2023; 102:587-605. [PMID: 37070386 DOI: 10.1111/cbdd.14257] [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: 11/07/2022] [Revised: 02/25/2023] [Accepted: 04/06/2023] [Indexed: 04/19/2023]
Abstract
Twenty different infectious disorders induced by bacteria, viruses, and parasites are categorized as neglected tropical diseases (NTDs) by WHO. The severity of chagas disease remains a major concern in endemic areas and an emerging public health hazard in nonendemic countries. Trypanosoma cruzi, the etiological agent of this NTD, is mostly transmitted by triatomine vectors and comprises a range of epidemiologically significant variants. Current chemotherapeutics are obsolete, and one of the primary reasons for treatment cessation is their poor safety and effectiveness. Due to the aforementioned challenges, researchers are now focusing on discovering alternative novel safe, and economically reachable therapies for the treatment of trypanosomiasis. Certain target-based drugs that target specific biochemical processes of the causative parasites have been described as potential antichagasic agents that possesses various types of heterocyclic scaffolds. These flexible molecules have a wide range of biological actions, and various synthesized compounds with strong activity have been documented. This review aims to discuss the available literature on synthetic anti-T. cruzi drugs that will give a food for thought to medicinal chemists thriving to design and develop such drugs. Furthermore, some of the studies discussed herein are concerned with the potential of novel drugs to block new viable sites in T. cruzi.
Collapse
Affiliation(s)
- Shilpi Pathak
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Muskan Bhardwaj
- Hospital Administration, FCAM, SGT University, Gurugram, India
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Aditya Bhardwaj
- Department of Healthcare Management, Chitkara Business School, Chitkara University, Punjab, India
| |
Collapse
|
14
|
Varela MT, Romanelli M, Amaral M, Tempone AG, Fernandes JPS. Piperazine amides with desirable solubility, physicochemical and drug-like properties: Synthesis and evaluation of the anti- Trypanosoma cruzi activity. Saudi Pharm J 2023; 31:1265-1273. [PMID: 37287509 PMCID: PMC10242637 DOI: 10.1016/j.jsps.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
The absence of effective chronic treatment, expansion to non-endemic countries and the significant burden in public health have stimulated the search for novel therapeutic options to treat Chagas disease, a protozoan disease caused by Trypanosoma cruzi. Despite current efforts, no new drug candidates were approved in clinical trials in the past five decades. Considering this, our group has focused on the expansion of a series (LINS03) with low micromolar activity against amastigotes, considering the optimization of pharmacokinetic properties through increasing drug-likeness and solubility. In this work, we report a new set of 13 compounds with modifications in both the arylpiperazine and the aromatic region linked by an amide group. Five analogues showed activity against intracellular amastigotes (IC50 17.8 to 35.9 µM) and no relevant cytotoxicity to mammalian cells (CC50 > 200 µM). Principal component analysis (PCA) was performed to identify structural features associated to improved activity. The data revealed that polarity, hydrogen bonding ability and flexibility were key properties that influenced the antiparasitic activity. In silico drug-likeness assessments indicated that compounds with the 4-methoxycinammyl (especially compound 2b) had the most prominent balance between properties and activity in the series, as confirmed by SAR analysis.
Collapse
Affiliation(s)
- Marina T. Varela
- Departamento de Medicina, Universidade Federal de São Paulo, Rua Botucatu 740, 04023-062 São Paulo, SP, Brazil
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo, Rua São Nicolau 210, 09913-030 Diadema, SP, Brazil
| | - Maiara Romanelli
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, Av. Dr. Arnaldo 351, 01246-000 São Paulo, SP, Brazil
| | - Maiara Amaral
- Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Andre G. Tempone
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, Av. Dr. Arnaldo 351, 01246-000 São Paulo, SP, Brazil
| | - João Paulo S. Fernandes
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo, Rua São Nicolau 210, 09913-030 Diadema, SP, Brazil
| |
Collapse
|
15
|
Nascimento IJDS, Cavalcanti MDAT, de Moura RO. Exploring N-myristoyltransferase as a promising drug target against parasitic neglected tropical diseases. Eur J Med Chem 2023; 258:115550. [PMID: 37336067 DOI: 10.1016/j.ejmech.2023.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Neglected tropical diseases (NTDs) constitute a group of approximately 20 infectious diseases that mainly affect the impoverished population without basic sanitation in tropical countries. These diseases are responsible for many deaths worldwide, costing billions of dollars in public health investment to treat and control these infections. Among them are the diseases caused by protozoa of the Trypanosomatid family, which constitute Trypanosoma cruzi (Chagas disease), Trypanosoma brucei (sleeping sickness), and Leishmaniasis. In addition, there is a classification of other diseases, called the big three, AIDS, tuberculosis, and malaria, which are endemic in countries with tropical conditions. Despite the high mortality rates, there is still a gap in the treatment. The drugs have a high incidence of side effects and protozoan resistance, justifying the investment in developing new alternatives. In fact, the Target-Based Drug Design (TBDD) approach is responsible for identifying several promising compounds, and among the targets explored through this approach, N-myristoyltransferase (NMT) stands out. It is an enzyme related to the co-translational myristoylation of N-terminal glycine in various peptides. The myristoylation process is a co-translation that occurs after removing the initiator methionine. This process regulates the assembly of protein complexes and stability, which justifies its potential as a drug target. In order to propose NMT as a potential target for parasitic diseases, this review will address the entire structure and function of this enzyme and the primary studies demonstrating its promising potential against Leishmaniasis, T. cruzi, T. brucei, and malaria. We hope our information can help researchers worldwide search for potential drugs against these diseases that have been threatening the health of the world's population.
Collapse
Affiliation(s)
- Igor José Dos Santos Nascimento
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil; Cesmac University Center, Pharmacy Departament, Maceió, Brazil; Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil.
| | - Misael de Azevedo Teotônio Cavalcanti
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil; Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Ricardo Olimpio de Moura
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil; Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| |
Collapse
|
16
|
Breslin W, Pham D. Machine learning and drug discovery for neglected tropical diseases. BMC Bioinformatics 2023; 24:165. [PMID: 37095460 PMCID: PMC10127295 DOI: 10.1186/s12859-022-05076-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/23/2022] [Indexed: 04/26/2023] Open
Abstract
Neglected tropical diseases affect millions of individuals and cause loss of productivity worldwide. They are common in developing countries without the financial resources for research and drug development. With increased availability of data from high throughput screening, machine learning has been introduced into the drug discovery process. Models can be trained to predict biological activities of compounds before working in the lab. In this study, we use three publicly available, high-throughput screening datasets to train machine learning models to predict biological activities related to inhibition of species that cause leishmaniasis, American trypanosomiasis (Chagas disease), and African trypanosomiasis (sleeping sickness). We compare machine learning models (tree based models, naive Bayes classifiers, and neural networks), featurizing methods (circular fingerprints, MACCS fingerprints, and RDKit descriptors), and techniques to deal with the imbalanced data (oversampling, undersampling, class weight/sample weight).
Collapse
Affiliation(s)
- William Breslin
- Department of Mathematics, Computer Science, and Data Science, Pacific University, Forest Grove, OR, USA.
| | - Doan Pham
- Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Hanover, NH, USA
| |
Collapse
|
17
|
Chen S, Li Z, Hu K, Feng W, Mao G, Xiao F, Deng GJ. Three-component selective synthesis of phenothiazines and bis-phenothiazines under metal-free conditions. Org Biomol Chem 2023; 21:1920-1926. [PMID: 36752306 DOI: 10.1039/d3ob00055a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
An iodine-containing reagent promoted three-component method for the selective synthesis of phenothiazines and bis-phenothiazines has been developed. The present protocol starts from simple and easily available cyclohexanones, elemental sulfur, and inorganic ammonium salts, selectively producing phenothiazines and bis-phenothiazines in satisfactory yields under aerobic conditions. This method has the advantages of simple and readily available starting materials and metal-free conditions, affording a facile and practical approach for the preparation of phenothiazines and bis-phenothiazines.
Collapse
Affiliation(s)
- Shanping Chen
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, P. R. China.
| | - Zhuoqin Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, P. R. China.
| | - Kai Hu
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, P. R. China.
| | - Wei Feng
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, P. R. China.
| | - Guojiang Mao
- School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang, 453007, P. R. China
| | - Fuhong Xiao
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, P. R. China.
| | - Guo-Jun Deng
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, P. R. China.
| |
Collapse
|
18
|
Guerra F, Winzeler EA. New targets for antimalarial drug discovery. Curr Opin Microbiol 2022; 70:102220. [PMID: 36228458 PMCID: PMC9934905 DOI: 10.1016/j.mib.2022.102220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/22/2022] [Accepted: 09/10/2022] [Indexed: 01/25/2023]
Abstract
Phenotypic screening methods have placed numerous preclinical candidates into the antimalarial drug-discovery pipeline. As more chemically validated targets become available, efforts are shifting to target-based drug discovery. Here, we briefly review some of the most attractive targets that have been identified in recent years.
Collapse
Affiliation(s)
- Francisco Guerra
- Department of Pediatrics MC 0760, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics MC 0760, School of Medicine, University of California, La Jolla, San Diego, CA 92093, USA.
| |
Collapse
|
19
|
Hierarchical Clustering and Target-Independent QSAR for Antileishmanial Oxazole and Oxadiazole Derivatives. Int J Mol Sci 2022; 23:ijms23168898. [PMID: 36012163 PMCID: PMC9408707 DOI: 10.3390/ijms23168898] [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: 06/22/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis is a neglected tropical disease that kills more than 20,000 people each year. The chemotherapy available for the treatment of the disease is limited, and novel approaches to discover novel drugs are urgently needed. Herein, 2D- and 4D-quantitative structure–activity relationship (QSAR) models were developed for a series of oxazole and oxadiazole derivatives that are active against Leishmania infantum, the causative agent of visceral leishmaniasis. A clustering strategy based on structural similarity was applied with molecular fingerprints to divide the complete set of compounds into two groups. Hierarchical clustering was followed by the development of 2D- (R2 = 0.90, R2pred = 0.82) and 4D-QSAR models (R2 = 0.80, R2pred = 0.64), which showed improved statistical robustness and predictive ability.
Collapse
|
20
|
Pöhner I, Quotadamo A, Panecka-Hofman J, Luciani R, Santucci M, Linciano P, Landi G, Di Pisa F, Dello Iacono L, Pozzi C, Mangani S, Gul S, Witt G, Ellinger B, Kuzikov M, Santarem N, Cordeiro-da-Silva A, Costi MP, Venturelli A, Wade RC. Multitarget, Selective Compound Design Yields Potent Inhibitors of a Kinetoplastid Pteridine Reductase 1. J Med Chem 2022; 65:9011-9033. [PMID: 35675511 PMCID: PMC9289884 DOI: 10.1021/acs.jmedchem.2c00232] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
The optimization
of compounds with multiple targets is a difficult
multidimensional problem in the drug discovery cycle. Here, we present
a systematic, multidisciplinary approach to the development of selective
antiparasitic compounds. Computational fragment-based design of novel
pteridine derivatives along with iterations of crystallographic structure
determination allowed for the derivation of a structure–activity
relationship for multitarget inhibition. The approach yielded compounds
showing apparent picomolar inhibition of T. brucei pteridine reductase 1 (PTR1), nanomolar inhibition of L.
major PTR1, and selective submicromolar inhibition of parasite
dihydrofolate reductase (DHFR) versus human DHFR. Moreover, by combining
design for polypharmacology with a property-based on-parasite optimization,
we found three compounds that exhibited micromolar EC50 values against T. brucei brucei while retaining
their target inhibition. Our results provide a basis for the further
development of pteridine-based compounds, and we expect our multitarget
approach to be generally applicable to the design and optimization
of anti-infective agents.
Collapse
Affiliation(s)
- Ina Pöhner
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), D-69118 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, D-69120 Heidelberg, Germany
| | - Antonio Quotadamo
- Tydock Pharma srl, Strada Gherbella 294/B, 41126 Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Joanna Panecka-Hofman
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), D-69118 Heidelberg, Germany.,Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | - Rosaria Luciani
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Matteo Santucci
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Pasquale Linciano
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Giacomo Landi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Flavio Di Pisa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Lucia Dello Iacono
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Nuno Santarem
- Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal.,Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria P Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Alberto Venturelli
- Tydock Pharma srl, Strada Gherbella 294/B, 41126 Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), D-69118 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, D-69120 Heidelberg, Germany.,Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, D-69120 Heidelberg, Germany.,Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, D-69120 Heidelberg, Germany
| |
Collapse
|
21
|
Liu L, Wang H, Lin L, Gao Y, Niu X. Mulberrin inhibits Botrytis cinerea for strawberry storage by interfering with the bioactivity of 14α-demethylase (CYP51). Food Funct 2022; 13:4032-4046. [PMID: 35315482 DOI: 10.1039/d2fo00295g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Currently, chemical agents hold great promise in preventing and combating Botrytis cinerea. However, the antifungal mechanism of some agents for B. cinerea remains rather vague, imposing restrictions on the research and development of novel antifungal inhibitors. In this work, we discovered that mulberrin (MBN), a natural compound from the root bark of Ramulus Mori, with an IC50 of 1.38 μM together, demonstrated marked anti-14α-demethylase (CYP51) activity through high throughput virtual screening and in vitro bioactivity assay. The computational biology results demonstrated that MBN and its derivatives were bound to the catalytic activity region of CYP51, but only MBN could form a strong π-cation interaction with the Fe ion of heme in CYP51 via the 2-methylpent-2-ene moiety at atom C9. MBN had a stronger binding free energy than the other three compounds with CYP51, implying that the 2-methylpent-2-ene moiety at atom C9 is a critical pharmacophore for CYP51 inhibitors. Subsequently, through an antifungal test, MBN demonstrated excellent anti-B. cinerea activity by inhibiting CYP51 activity. The EC50 values of MBN toward hyphal growth and spore germination in B. cinerea were 17.27 and 9.56 μg mL-1, respectively. The bioactivity loss of CYP51 by direct interaction with MBN induced the increase of cell membrane permeability, membrane destruction, and cell death. Meanwhile, in the B. cinerea infection model, MBN significantly prolonged the preservation of strawberries by preventing B. cinerea from infecting strawberries and could be used as a potential natural preserving agent for storing fruits.
Collapse
Affiliation(s)
- Lu Liu
- College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Hongsu Wang
- College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Li Lin
- College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Yawen Gao
- College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Xiaodi Niu
- College of Food Science and Engineering, Jilin University, Changchun, China.
| |
Collapse
|
22
|
Varela MT, Amaral M, Romanelli MM, de Castro Levatti EV, Tempone AG, Fernandes JPS. Optimization of physicochemical properties is a strategy to improve drug-likeness associated with activity: novel active and selective compounds against Trypanosoma cruzi. Eur J Pharm Sci 2022; 171:106114. [PMID: 34986415 DOI: 10.1016/j.ejps.2021.106114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/23/2021] [Accepted: 12/31/2021] [Indexed: 11/03/2022]
Abstract
Trypanosoma cruzi is the causing agent of Chagas disease, a parasitic infection without efficient treatment for chronic patients. Despite the efforts, no new drugs have been approved for this disease in the last 60 years. Molecular modifications based on a natural product led to the development of a series of compounds (LINS03 series) with promising antitrypanosomal activity, however previous chemometric analysis revealed a significant impact of excessive lipophilicity and low aqueous solubility on potency of amine and amide derivatives. Therefore, this work reports different modifications in the core structure to achieve adequate balance of the physicochemical properties along with biological activity. A set of 34 analogues were designed considering predicted properties related to lipophilicity/hydrosolubility and synthesized to assess their activity and selective toxicity towards the parasite. Results showed that this strategy contributed to improve the drug-likeness of the series while considerable impacts on potency were observed. The rational analysis of the obtained data led to the identification of seven active piperazine amides (28-34, IC50 8.7 to 35.3 µM against intracellular amastigotes), devoid of significant cytotoxicity to mammalian cells. The addition of water-solubilizing groups and privileged substructures such as piperazines improved the physicochemical properties and overall drug-likeness of these compounds, increased potency and maintained selectivity towards the parasite. The obtained results brought important structure-activity relationship (SAR) data and new lead structures for further modifications were identified to achieve improved antitrypanosoma compounds.
Collapse
Affiliation(s)
- Marina T Varela
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo, Rua São Nicolau 210, 09913-030 Diadema SP, Brazil
| | - Maiara Amaral
- Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Maiara M Romanelli
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, Av. Dr. Arnaldo 351, 01246-000 São Paulo SP, Brazil
| | - Erica V de Castro Levatti
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, Av. Dr. Arnaldo 351, 01246-000 São Paulo SP, Brazil
| | - Andre G Tempone
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, Av. Dr. Arnaldo 351, 01246-000 São Paulo SP, Brazil
| | - João Paulo S Fernandes
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo, Rua São Nicolau 210, 09913-030 Diadema SP, Brazil.
| |
Collapse
|
23
|
A novel high-content phenotypic screen to identify inhibitors of mitochondrial DNA maintenance in trypanosomes. Antimicrob Agents Chemother 2021; 66:e0198021. [PMID: 34871097 PMCID: PMC8846439 DOI: 10.1128/aac.01980-21] [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] [Indexed: 11/30/2022] Open
Abstract
Kinetoplastid parasites cause diverse neglected diseases in humans and livestock, with an urgent need for new treatments. The survival of kinetoplastids depends on their uniquely structured mitochondrial genome (kDNA), the eponymous kinetoplast. Here, we report the development of a high-content screen for pharmacologically induced kDNA loss, based on specific staining of parasites and automated image analysis. As proof of concept, we screened a diverse set of ∼14,000 small molecules and exemplify a validated hit as a novel kDNA-targeting compound.
Collapse
|
24
|
da Costa-Silva TA, Silva ML, Antar GM, Tempone AG, Lago JHG. Ent-kaurane diterpenes isolated from n-hexane extract of Baccharis sphenophylla by bioactivity-guided fractionation target the acidocalcisomes in Trypanosoma cruzi. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153748. [PMID: 34628240 DOI: 10.1016/j.phymed.2021.153748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND In the present work the bioactivity-guided fractionation of n-hexane extract from aerial parts of Baccharis sphenophylla (Asteraceae) against trypomastigote forms of Trypanosoma cruzi was performed. PURPOSE To evaluate the antitrypanosomal potential of diterpenes ent‑kaurenoic (1), grandifloric (2). and 15β-tiglinoyloxy‑ent-kaurenoic (3) acids, isolated from n-hexane extract from aerial parts of B. sphenophylla, and elucidate their mechanism of action against T. cruzi. METHODS/STUDY DESIGN: n-Hexane and MeOH extracts from aerial parts of B. sphenophylla were prepared and caused, respectively, 100% and 50% of death of trypomastigote forms of T. cruzi. Based on these results, the n-hexane extract was subjected to bioactivity-guided fractionation procedures to afford three related ent‑kaurane diterpenoids (1-3). Based on spectrofluorometric assays and flow cytometry analysis, the mechanism of action of compounds 1 and 3 was investigated. RESULTS Compounds 1 and 3, isolated from n-hexane extract from aerial parts of B. sphenophylla, showed potent activity against parasites with EC50 values of 10.6 μM (SI > 18.8) and 2.4 μM (SI = 34.8), respectively. On the other hand, compound 2 was inactive against trypomastigotes. In mechanism of action studies using the fluorescent probe SYTOX Green, the plasma membrane permeability was unaltered after treatment with compounds 1 and 3, but compound 1 induced a depolarization of the plasma membrane electric potential (ΔΨp). No substantial alterations were observed in the mitochondria after treatment with compound 3, but a transient hyperpolarization of the mitochondrial membrane potential (ΔΨm) by compound 1. Despite the increased ATP levels induced by compounds 1 and 3, no alterations of ROS and Ca2+ levels were registered. However, both compounds promoted a time-dependent alkalinization of the acidocalcisomes, probably contributing to an osmotic imbalance of the cell. In silico physicochemical studies of compounds 1-3 suggested that lipophilicity and molecular complexity may play an important role in the antitrypanosomal activity. Moreover, no pan-assay interference compounds (PAINS) alerts were detected for compounds 1-3. CONCLUSION Obtained data indicated that the isolated ent‑kaurane diterpenes from n-hexane extract from aerial parts of B. sphenophylla, especially compound 3, could be considered interesting prototypes for further modifications aiming the discovery of new hits against T. cruzi.
Collapse
Affiliation(s)
- Thais A da Costa-Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, São Paulo 09210-180, Brazil
| | - Matheus L Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, São Paulo 09210-180, Brazil
| | - Guilherme M Antar
- Department of Botany, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Andre G Tempone
- Center for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo 01246-902, Brazil.
| | - João Henrique G Lago
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, São Paulo 09210-180, Brazil.
| |
Collapse
|
25
|
Herrera-Acevedo C, Dos Santos Maia M, Cavalcanti ÉBVS, Coy-Barrera E, Scotti L, Scotti MT. Selection of antileishmanial sesquiterpene lactones from SistematX database using a combined ligand-/structure-based virtual screening approach. Mol Divers 2021; 25:2411-2427. [PMID: 32909084 DOI: 10.1007/s11030-020-10139-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/30/2020] [Indexed: 12/20/2022]
Abstract
Leishmaniasis refers to a complex of diseases, caused by the intracellular parasitic protozoans belonging to the genus Leishmania. Among the three types of disease manifestations, the most severe type is visceral leishmaniasis, which is caused by Leishmania donovani, and is diagnosed in more than 20,000 cases annually, worldwide. Because the current therapeutic options for disease treatment are associated with several limitations, the identification of new potential leads/drugs remains necessary. In this study, a combined approach was used, based on two different virtual screening (VS) methods, which were designed to select promising antileishmanial agents from among the entire sesquiterpene lactone (SL) dataset registered in SistematX, a web interface for managing a secondary metabolite database that is accessible by multiple platforms on the Internet. Thus, a ChEMBL dataset, including 3159 and 1569 structures that were previously tested against L. donovani amastigotes and promastigotes in vitro, respectively, was used to develop two random forest models, which performed with greater than 74% accuracy in both the cross-validation and test sets. Subsequently, a ligand-based VS assay was performed against the 1306 SistematX-registered SLs. In parallel, the crystal structures of three L. donovani target proteins, N-myristoyltransferase, ornithine decarboxylase, and mitogen-activated protein kinase 3, and a homology model of pteridine reductase 1 were used to perform a structure-based VS, using molecular docking, of the entire SistematX SL dataset. The consensus analysis of these two VS approaches resulted in the normalization of probability scores and identified 13 promising, enzyme-targeting, antileishmanial SLs from SistematX that may act against L. donovani. A combined approach based on two different virtual screening methods (structure-based and ligand-based) was performed using an in-house dataset composed of 1306 sesquiterpene lactones to identify potential antileishmanial (Leishmania donovani) structures.
Collapse
Affiliation(s)
- Chonny Herrera-Acevedo
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, 58051-900, Brazil
- Bioorganic Chemistry Laboratory, Facultad de Ciencias Básicas y Aplicadas, Universidad Militar Nueva Granada, Cajicá, 250247, Colombia
| | - Mayara Dos Santos Maia
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, 58051-900, Brazil
| | | | - Ericsson Coy-Barrera
- Bioorganic Chemistry Laboratory, Facultad de Ciencias Básicas y Aplicadas, Universidad Militar Nueva Granada, Cajicá, 250247, Colombia
| | - Luciana Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, 58051-900, Brazil
| | - Marcus Tullius Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, 58051-900, Brazil.
| |
Collapse
|
26
|
de Oliveira RG, Cruz LR, Mollo MC, Dias LC, Kratz JM. Chagas Disease Drug Discovery in Latin America-A Mini Review of Antiparasitic Agents Explored Between 2010 and 2021. Front Chem 2021; 9:771143. [PMID: 34778217 PMCID: PMC8581468 DOI: 10.3389/fchem.2021.771143] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi that endangers almost 70 million people worldwide. The only two drugs that are currently approved for its treatment, benznidazole and nifurtimox, have controversial efficacy in adults and restricting safety issues, leaving thousands of patients without a suitable treatment. The neglect of Chagas disease is further illustrated by the lack of a robust and diverse drug discovery and development portfolio of new chemical entities, and it is of paramount importance to build a strong research and development network for antichagasic drugs. Focusing on drug discovery programs led by scientists based in Latin America, the main endemic region for this disease, we discuss herein what has been published in the last decade in terms of identification of new antiparasitic drugs to treat Chagas disease, shining a spotlight on the origin, chemical diversity, level of characterization of hits, and strategies used for optimization of lead compounds. Finally, we identify strengths and weaknesses in these drug discovery campaigns and highlight the importance of multidisciplinary collaboration and knowledge sharing.
Collapse
Affiliation(s)
- Ramon G. de Oliveira
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, Brazil
| | - Luiza R. Cruz
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, Brazil
| | - María C. Mollo
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, Brazil
| | - Luiz C. Dias
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, Brazil
| | - Jadel M. Kratz
- Drugs for Neglected Diseases Initiative (DNDi) Latin America, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Forte B, Ottilie S, Plater A, Campo B, Dechering KJ, Gamo FJ, Goldberg DE, Istvan ES, Lee M, Lukens AK, McNamara CW, Niles JC, Okombo J, Pasaje CFA, Siegel MG, Wirth D, Wyllie S, Fidock DA, Baragaña B, Winzeler EA, Gilbert IH. Prioritization of Molecular Targets for Antimalarial Drug Discovery. ACS Infect Dis 2021; 7:2764-2776. [PMID: 34523908 PMCID: PMC8608365 DOI: 10.1021/acsinfecdis.1c00322] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
There is a shift
in antimalarial drug discovery from phenotypic
screening toward target-based approaches, as more potential drug targets
are being validated in Plasmodium species. Given
the high attrition rate and high cost of drug discovery, it is important
to select the targets most likely to deliver progressible drug candidates.
In this paper, we describe the criteria that we consider important
for selecting targets for antimalarial drug discovery. We describe
the analysis of a number of drug targets in the Malaria Drug Accelerator
(MalDA) pipeline, which has allowed us to prioritize targets that
are ready to enter the drug discovery process. This selection process
has also highlighted where additional data are required to inform
target progression or deprioritization of other targets. Finally,
we comment on how additional drug targets may be identified.
Collapse
Affiliation(s)
- Barbara Forte
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Andrew Plater
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Brice Campo
- Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | | | | | - Daniel E. Goldberg
- Division of Infectious Diseases, Department of Medicine and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Eva S. Istvan
- Division of Infectious Diseases, Department of Medicine and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Marcus Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, United Kingdom
| | - Amanda K. Lukens
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Case W. McNamara
- Calibr, a Division of The Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Jacquin C. Niles
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge Massachusetts 02139-4307, United States
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Charisse Flerida A. Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge Massachusetts 02139-4307, United States
| | | | - Dyann Wirth
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Beatriz Baragaña
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Elizabeth A. Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Ian H. Gilbert
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, DD1 5EH, United Kingdom
| |
Collapse
|
28
|
Lande DH, Nasereddin A, Alder A, Gilberger TW, Dzikowski R, Grünefeld J, Kunick C. Synthesis and Antiplasmodial Activity of Bisindolylcyclobutenediones. Molecules 2021; 26:4739. [PMID: 34443327 PMCID: PMC8402075 DOI: 10.3390/molecules26164739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/02/2022] Open
Abstract
Malaria is one of the most dangerous infectious diseases. Because the causative Plasmodium parasites have developed resistances against virtually all established antimalarial drugs, novel antiplasmodial agents are required. In order to target plasmodial kinases, novel N-unsubstituted bisindolylcyclobutenediones were designed as analogs to the kinase inhibitory bisindolylmaleimides. Molecular docking experiments produced favorable poses of the unsubstituted bisindolylcyclobutenedione in the ATP binding pocket of various plasmodial protein kinases. The synthesis of the title compounds was accomplished by sequential Friedel-Crafts acylation procedures. In vitro screening of the new compounds against transgenic NF54-luc P. falciparum parasites revealed a set of derivatives with submicromolar activity, of which some displayed a reasonable selectivity profile against a human cell line. Although the molecular docking studies suggested the plasmodial protein kinase PfGSK-3 as the putative biological target, the title compounds failed to inhibit the isolated enzyme in vitro. As selective submicromolar antiplasmodial agents, the N-unsubstituted bisindolylcyclobutenediones are promising starting structures in the search for antimalarial drugs, albeit for a rational development, the biological target addressed by these compounds has yet to be identified.
Collapse
Affiliation(s)
- Duc Hoàng Lande
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethoven straße 55, 38106 Braunschweig, Germany; (D.H.L.); (J.G.)
| | - Abed Nasereddin
- Department of Microbiology and Molecular Genetics, IMRIC, The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel; (A.N.); (R.D.)
- Genomics Applications Laboratory, Core Research Facility, Faculty of Medicine, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Arne Alder
- Centre for Structural Systems Biology, 22607 Hamburg, Germany; (A.A.); (T.W.G.)
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- Department of Biology, University of Hamburg, 20146 Hamburg, Germany
| | - Tim W. Gilberger
- Centre for Structural Systems Biology, 22607 Hamburg, Germany; (A.A.); (T.W.G.)
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- Department of Biology, University of Hamburg, 20146 Hamburg, Germany
| | - Ron Dzikowski
- Department of Microbiology and Molecular Genetics, IMRIC, The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel; (A.N.); (R.D.)
| | - Johann Grünefeld
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethoven straße 55, 38106 Braunschweig, Germany; (D.H.L.); (J.G.)
| | - Conrad Kunick
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethoven straße 55, 38106 Braunschweig, Germany; (D.H.L.); (J.G.)
- Zentrum für Pharmaverfahrenstechnik (PVZ), Technische Universität Braunschweig, Franz-Liszt-Straße 35A, 38106 Braunschweig, Germany
| |
Collapse
|
29
|
Morales JF, Chuguransky S, Alberca LN, Alice JI, Goicoechea S, Ruiz ME, Bellera CL, Talevi A. Positive Predictive Value Surfaces as a Complementary Tool to Assess the Performance of Virtual Screening Methods. Mini Rev Med Chem 2021; 20:1447-1460. [PMID: 32072906 DOI: 10.2174/1871525718666200219130229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Since their introduction in the virtual screening field, Receiver Operating Characteristic (ROC) curve-derived metrics have been widely used for benchmarking of computational methods and algorithms intended for virtual screening applications. Whereas in classification problems, the ratio between sensitivity and specificity for a given score value is very informative, a practical concern in virtual screening campaigns is to predict the actual probability that a predicted hit will prove truly active when submitted to experimental testing (in other words, the Positive Predictive Value - PPV). Estimation of such probability is however, obstructed due to its dependency on the yield of actives of the screened library, which cannot be known a priori. OBJECTIVE To explore the use of PPV surfaces derived from simulated ranking experiments (retrospective virtual screening) as a complementary tool to ROC curves, for both benchmarking and optimization of score cutoff values. METHODS The utility of the proposed approach is assessed in retrospective virtual screening experiments with four datasets used to infer QSAR classifiers: inhibitors of Trypanosoma cruzi trypanothione synthetase; inhibitors of Trypanosoma brucei N-myristoyltransferase; inhibitors of GABA transaminase and anticonvulsant activity in the 6 Hz seizure model. RESULTS Besides illustrating the utility of PPV surfaces to compare the performance of machine learning models for virtual screening applications and to select an adequate score threshold, our results also suggest that ensemble learning provides models with better predictivity and more robust behavior. CONCLUSION PPV surfaces are valuable tools to assess virtual screening tools and choose score thresholds to be applied in prospective in silico screens. Ensemble learning approaches seem to consistently lead to improved predictivity and robustness.
Collapse
Affiliation(s)
- Juan F Morales
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - Sara Chuguransky
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - Lucas N Alberca
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - Juan I Alice
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - Sofía Goicoechea
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - María E Ruiz
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - Carolina L Bellera
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| | - Alan Talevi
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata (UNLP) - 47 & 115, La Plata (1900), Buenos Aires, Argentina
| |
Collapse
|
30
|
SARs for the Antiparasitic Plant Metabolite Pulchrol. 3. Combinations of New Substituents in A/B-Rings and A/C-Rings. Molecules 2021; 26:molecules26133944. [PMID: 34203527 PMCID: PMC8271509 DOI: 10.3390/molecules26133944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
The natural products pulchrol and pulchral, isolated from the roots of the Mexican plant Bourreria pulchra, have previously been shown to possess antiparasitic activity towards Trypanosoma cruzi, Leishmania braziliensis and L. amazonensis, which are protozoa responsible for Chagas disease and leishmaniasis. These infections have been classified as neglected diseases, and still require the development of safer and more efficient alternatives to their current treatments. Recent SARs studies, based on the pulchrol scaffold, showed which effects exchanges of its substituents have on the antileishmanial and antitrypanosomal activity. Many of the analogues prepared were shown to be more potent than pulchrol and the current drugs used to treat leishmaniasis and Chagas disease (miltefosine and benznidazole, respectively), in vitro. Moreover, indications of some of the possible interactions that may take place in the binding sites were also identified. In this study, 12 analogues with modifications at two or three different positions in two of the three rings were prepared by synthetic and semi-synthetic procedures. The molecules were assayed in vitro towards T. cruzi epimastigotes, L. braziliensis promastigotes, and L. amazonensis promastigotes. Some compounds had higher antiparasitic activity than the parental compound pulchrol, and in some cases even benznidazole and miltefosine. The best combinations in this subset are with carbonyl functionalities in the A-ring and isopropyl groups in the C-ring, as well as with alkyl substituents in both the A- and C-rings combined with a hydroxyl group in position 1 (C-ring). The latter corresponds to cannabinol, which indeed was shown to be potent towards all the parasites.
Collapse
|
31
|
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: 2.0] [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
|
32
|
dos Santos Vasconcelos CR, Rezende AM. Systematic in silico Evaluation of Leishmania spp. Proteomes for Drug Discovery. Front Chem 2021; 9:607139. [PMID: 33987166 PMCID: PMC8111926 DOI: 10.3389/fchem.2021.607139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/24/2021] [Indexed: 11/18/2022] Open
Abstract
Leishmaniasis is a group of neglected infectious diseases, with approximately 1. 3 million new cases each year, for which the available therapies have serious limitations. Therefore, it is extremely important to apply efficient and low-cost methods capable of selecting the best therapeutic targets to speed up the development of new therapies against those diseases. Thus, we propose the use of integrated computational methods capable of evaluating the druggability of the predicted proteomes of Leishmania braziliensis and Leishmania infantum, species responsible for the different clinical manifestations of leishmaniasis in Brazil. The protein members of those proteomes were assessed based on their structural, chemical, and functional contexts applying methods that integrate data on molecular function, biological processes, subcellular localization, drug binding sites, druggability, and gene expression. These data were compared to those extracted from already known drug targets (BindingDB targets), which made it possible to evaluate Leishmania proteomes for their biological relevance and treatability. Through this methodology, we identified more than 100 proteins of each Leishmania species with druggability characteristics, and potential interaction with available drugs. Among those, 31 and 37 proteins of L. braziliensis and L. infantum, respectively, have never been tested as drug targets, and they have shown evidence of gene expression in the evolutionary stage of pharmacological interest. Also, some of those Leishmania targets showed an alignment similarity of <50% when compared to the human proteome, making these proteins pharmacologically attractive, as they present a reduced risk of side effects. The methodology used in this study also allowed the evaluation of opportunities for the repurposing of compounds as anti-leishmaniasis drugs, inferring potential interaction between Leishmania proteins and ~1,000 compounds, of which only 15 have already been tested as a treatment for leishmaniasis. Besides, a list of potential Leishmania targets to be tested using drugs described at BindingDB, such as the potential interaction of the DEAD box RNA helicase, TRYR, and PEPCK proteins with the Staurosporine compound, was made available to the public.
Collapse
Affiliation(s)
- Crhisllane Rafaele dos Santos Vasconcelos
- Bioinformatics Plataform, Microbiology Department, Instituto Aggeu Magalhães, Recife, Brazil
- Posgraduate Program in Genetics, Genetics Department, Universidade Federal de Pernambuco, Recife, Brazil
| | - Antonio Mauro Rezende
- Bioinformatics Plataform, Microbiology Department, Instituto Aggeu Magalhães, Recife, Brazil
- Posgraduate Program in Genetics, Genetics Department, Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
33
|
Bobrova AV, Krasnov PO, Povarov IG, Bobrov PS, Lyubyashkin AV, Suboch GA, Tovbis MS. Facile synthesis and sulfonylation of 4-aminopyrazoles. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.129912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
34
|
Drug screening to identify compounds to act as co-therapies for the treatment of Burkholderia species. PLoS One 2021; 16:e0248119. [PMID: 33764972 PMCID: PMC7993816 DOI: 10.1371/journal.pone.0248119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/21/2021] [Indexed: 11/22/2022] Open
Abstract
Burkholderia pseudomallei is a soil-dwelling organism present throughout the tropics. It is the causative agent of melioidosis, a disease that is believed to kill 89,000 people per year. It is naturally resistant to many antibiotics, requiring at least two weeks of intravenous treatment with ceftazidime, imipenem or meropenem followed by 6 months of orally delivered co-trimoxazole. This places a large treatment burden on the predominantly middle-income nations where the majority of disease occurs. We have established a high-throughput assay for compounds that could be used as a co-therapy to potentiate the effect of ceftazidime, using the related non-pathogenic bacterium Burkholderia thailandensis as a surrogate. Optimization of the assay gave a Z’ factor of 0.68. We screened a library of 61,250 compounds and identified 29 compounds with a pIC50 (-log10(IC50)) greater than five. Detailed investigation allowed us to down select to six “best in class” compounds, which included the licensed drug chloroxine. Co-treatment of B. thailandensis with ceftazidime and chloroxine reduced culturable cell numbers by two orders of magnitude over 48 hours, compared to treatment with ceftazidime alone. Hit expansion around chloroxine was performed using commercially available compounds. Minor modifications to the structure abolished activity, suggesting that chloroxine likely acts against a specific target. Finally, an initial study demonstrates the utility of chloroxine to act as a co-therapy to potentiate the effect of ceftazidime against B. pseudomallei. This approach successfully identified potential co-therapies for a recalcitrant Gram-negative bacterial species. Our assay could be used more widely to aid in chemotherapy to treat infections caused by these bacteria.
Collapse
|
35
|
Arendse LB, Wyllie S, Chibale K, Gilbert IH. Plasmodium Kinases as Potential Drug Targets for Malaria: Challenges and Opportunities. ACS Infect Dis 2021; 7:518-534. [PMID: 33590753 PMCID: PMC7961706 DOI: 10.1021/acsinfecdis.0c00724] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Indexed: 12/30/2022]
Abstract
Protein and phosphoinositide kinases have been successfully exploited as drug targets in various disease areas, principally in oncology. In malaria, several protein kinases are under investigation as potential drug targets, and an inhibitor of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4KIIIβ) is currently in phase 2 clinical studies. In this Perspective, we review the potential of kinases as drug targets for the treatment of malaria. Kinases are known to be readily druggable, and many are essential for parasite survival. A key challenge in the design of Plasmodium kinase inhibitors is obtaining selectivity over the corresponding human orthologue(s) and other human kinases due to the highly conserved nature of the shared ATP binding site. Notwithstanding this, there are some notable differences between the Plasmodium and human kinome that may be exploitable. There is also the potential for designed polypharmacology, where several Plasmodium kinases are inhibited by the same drug. Prior to starting the drug discovery process, it is important to carefully assess potential kinase targets to ensure that the inhibition of the desired kinase will kill the parasites in the required life-cycle stages with a sufficiently fast rate of kill. Here, we highlight key target attributes and experimental approaches to consider and summarize the progress that has been made targeting Plasmodium PI4KIIIβ, cGMP-dependent protein kinase, and cyclin-dependent-like kinase 3.
Collapse
Affiliation(s)
- Lauren B. Arendse
- Drug
Discovery and Development Centre (H3D), South African Medical Research
Council Drug Discovery and Development Research Unit, Department of
Chemistry, and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Cape Town, Western Cape 7701, South Africa
| | - Susan Wyllie
- Wellcome
Centre for Anti-Infectives Research, Division of Biological Chemistry
and Drug Discovery, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Kelly Chibale
- Drug
Discovery and Development Centre (H3D), South African Medical Research
Council Drug Discovery and Development Research Unit, Department of
Chemistry, and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Cape Town, Western Cape 7701, South Africa
| | - Ian H. Gilbert
- Wellcome
Centre for Anti-Infectives Research, Division of Biological Chemistry
and Drug Discovery, University of Dundee, Dundee DD1 5EH, United Kingdom
| |
Collapse
|
36
|
J B, M BM, Chanda K. An Overview on the Therapeutics of Neglected Infectious Diseases-Leishmaniasis and Chagas Diseases. Front Chem 2021; 9:622286. [PMID: 33777895 PMCID: PMC7994601 DOI: 10.3389/fchem.2021.622286] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/14/2021] [Indexed: 12/20/2022] Open
Abstract
Neglected tropical diseases (NTDs) as termed by WHO include twenty different infectious diseases that are caused by bacteria, viruses, and parasites. Among these NTDs, Chagas disease and leishmaniasis are reported to cause high mortality in humans and are further associated with the limitations of existing drugs like severe toxicity and drug resistance. The above hitches have rendered researchers to focus on developing alternatives and novel therapeutics for the treatment of these diseases. In the past decade, several target-based drugs have emerged, which focus on specific biochemical pathways of the causative parasites. For leishmaniasis, the targets such as nucleoside analogs, inhibitors targeting nucleoside phosphate kinases of the parasite’s purine salvage pathway, 20S proteasome of Leishmania, mitochondria, and the associated proteins are reviewed along with the chemical structures of potential drug candidates. Similarly, in case of therapeutics for Chagas disease, several target-based drug candidates targeting sterol biosynthetic pathway (C14-ademethylase), L-cysteine protease, heme peroxidation, mitochondria, farnesyl pyrophosphate, etc., which are vital and unique to the causative parasite are discussed. Moreover, the use of nano-based formulations towards the therapeutics of the above diseases is also discussed.
Collapse
Affiliation(s)
- Brindha J
- Division of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Chennai, India
| | - Balamurali M M
- Division of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Chennai, India
| | - Kaushik Chanda
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
37
|
Alcazar W, Alakurtti S, Padrón-Nieves M, Tuononen ML, Rodríguez N, Yli-Kauhaluoma J, Ponte-Sucre A. Leishmanicidal Activity of Betulin Derivatives in Leishmania amazonensis; Effect on Plasma and Mitochondrial Membrane Potential, and Macrophage Nitric Oxide and Superoxide Production. Microorganisms 2021; 9:320. [PMID: 33557150 PMCID: PMC7913927 DOI: 10.3390/microorganisms9020320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 11/25/2022] Open
Abstract
Herein, we evaluated in vitro the anti-leishmanial activity of betulin derivatives in Venezuelan isolates of Leishmania amazonensis, isolated from patients with therapeutic failure. METHODS We analyzed promastigote in vitro susceptibility as well as the cytotoxicity and selectivity of the evaluated compounds. Additionally, the activity of selected compounds was determined in intracellular amastigotes. Finally, to gain hints on their potential mechanism of action, the effect of the most promising compounds on plasma and mitochondrial membrane potential, and nitric oxide and superoxide production by infected macrophages was determined. RESULTS From the tested 28 compounds, those numbered 18 and 22 were chosen for additional studies. Both 18 and 22 were active (GI50 ≤ 2 µM, cytotoxic CC50 > 45 µM, SI > 20) for the reference strain LTB0016 and for patient isolates. The results suggest that 18 significantly depolarized the plasma membrane potential (p < 0.05) and the mitochondrial membrane potential (p < 0.05) when compared to untreated cells. Although neither 18 nor 22 induced nitric oxide production in infected macrophages, 18 induced superoxide production in infected macrophages. CONCLUSION Our results suggest that due to their efficacy and selectivity against intracellular parasites and the potential mechanisms underlying their leishmanicidal effect, the compounds 18 and 22 could be used as tools for designing new chemotherapies against leishmaniasis.
Collapse
Affiliation(s)
- Wilmer Alcazar
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, P.O. Box 50587, Caracas 1050, Venezuela; (W.A.); (M.P.-N.)
| | - Sami Alakurtti
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, FI-00014 Helsinki, Finland; (S.A.); (M.L.T.)
- VTT Technical Research Centre of Finland, Biologinkuja 7, P.O. Box 1000, FI-02044 Espoo, Finland
| | - Maritza Padrón-Nieves
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, P.O. Box 50587, Caracas 1050, Venezuela; (W.A.); (M.P.-N.)
| | - Maija Liisa Tuononen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, FI-00014 Helsinki, Finland; (S.A.); (M.L.T.)
- VTT Technical Research Centre of Finland, Biologinkuja 7, P.O. Box 1000, FI-02044 Espoo, Finland
| | - Noris Rodríguez
- Laboratory of Genetic Engineering, Institute of Biomedicine, Universidad Central de Venezuela, P.O. Box 4043, Caracas 1010A, Venezuela;
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, FI-00014 Helsinki, Finland; (S.A.); (M.L.T.)
| | - Alicia Ponte-Sucre
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, P.O. Box 50587, Caracas 1050, Venezuela; (W.A.); (M.P.-N.)
| |
Collapse
|
38
|
Akao Y, Canan S, Cao Y, Condroski K, Engkvist O, Itono S, Kaki R, Kimura C, Kogej T, Nagaoka K, Naito A, Nakai H, Pairaudeau G, Radu C, Roberts I, Shimada M, Shum D, Watanabe NA, Xie H, Yonezawa S, Yoshida O, Yoshida R, Mowbray C, Perry B. Collaborative virtual screening to elaborate an imidazo[1,2- a]pyridine hit series for visceral leishmaniasis. RSC Med Chem 2021; 12:384-393. [PMID: 34041487 PMCID: PMC8130605 DOI: 10.1039/d0md00353k] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
An innovative pre-competitive virtual screening collaboration was engaged to validate and subsequently explore an imidazo[1,2-a]pyridine screening hit for visceral leishmaniasis. In silico probing of five proprietary pharmaceutical company libraries enabled rapid expansion of the hit chemotype, alleviating initial concerns about the core chemical structure while simultaneously improving antiparasitic activity and selectivity index relative to the background cell line. Subsequent hit optimization informed by the structure–activity relationship enabled by this virtual screening allowed thorough investigation of the pharmacophore, opening avenues for further improvement and optimization of the chemical series. Ligand-based similarity screening of proprietary pharmaceutical company libraries enables rapid hit to lead investigation of a chemotype with anti-leishmania activity.![]()
Collapse
Affiliation(s)
- Yuichiro Akao
- Takeda Pharmaceutical Company Limited 26-1 Muraoka-Higashi 2-chrome Fujisawa Kanagawa 251-8555 Japan
| | - Stacie Canan
- Celgene Corporation, Celgene Global Health 10300 Campus Point Drive San Diego California 92121 USA
| | - Yafeng Cao
- WuXi AppTec Company Ltd. 666 Gaoxin Road, East Lake High-Tech Development Zone Wuhan 430075 People's Republic of China
| | - Kevin Condroski
- Celgene Corporation, Celgene Global Health 10300 Campus Point Drive San Diego California 92121 USA
| | - Ola Engkvist
- AstraZeneca Discovery Sciences, R&D AstraZeneca Gothenburg Sweden
| | - Sachiko Itono
- Takeda Pharmaceutical Company Limited 26-1 Muraoka-Higashi 2-chrome Fujisawa Kanagawa 251-8555 Japan
| | - Rina Kaki
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Chiaki Kimura
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Thierry Kogej
- AstraZeneca Discovery Sciences, R&D AstraZeneca Gothenburg Sweden
| | - Kazuya Nagaoka
- Eisai Co., Ltd 1-3,Tokodai 5-chome Tsukuba Ibaraki 300-2635 Japan
| | - Akira Naito
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Hiromi Nakai
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | | | - Constantin Radu
- Institut Pasteur Korea 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu Seongnam-si Gyeonggi-do 13488 Republic of Korea
| | - Ieuan Roberts
- AstraZeneca, Discovery Sciences, R&D AstraZeneca Cambridge UK
| | - Mitsuyuki Shimada
- Takeda Pharmaceutical Company Limited 26-1 Muraoka-Higashi 2-chrome Fujisawa Kanagawa 251-8555 Japan
| | - David Shum
- Institut Pasteur Korea 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu Seongnam-si Gyeonggi-do 13488 Republic of Korea
| | - Nao-Aki Watanabe
- Eisai Co., Ltd 1-3,Tokodai 5-chome Tsukuba Ibaraki 300-2635 Japan
| | - Huanxu Xie
- WuXi AppTec Company Ltd. 666 Gaoxin Road, East Lake High-Tech Development Zone Wuhan 430075 People's Republic of China
| | - Shuji Yonezawa
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Osamu Yoshida
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Ryu Yoshida
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Charles Mowbray
- Drugs for Neglected Diseases initiative 15 Chemin Louis Dunant Geneva 1202 Switzerland
| | - Benjamin Perry
- Drugs for Neglected Diseases initiative 15 Chemin Louis Dunant Geneva 1202 Switzerland
| |
Collapse
|
39
|
Mansoldo FRP, Carta F, Angeli A, Cardoso VDS, Supuran CT, Vermelho AB. Chagas Disease: Perspectives on the Past and Present and Challenges in Drug Discovery. Molecules 2020; 25:E5483. [PMID: 33238613 PMCID: PMC7700143 DOI: 10.3390/molecules25225483] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
Chagas disease still has no effective treatment option for all of its phases despite being discovered more than 100 years ago. The development of commercial drugs has been stagnating since the 1960s, a fact that sheds light on the question of how drug discovery research has progressed and taken advantage of technological advances. Could it be that technological advances have not yet been sufficient to resolve this issue or is there a lack of protocol, validation and standardization of the data generated by different research teams? This work presents an overview of commercial drugs and those that have been evaluated in studies and clinical trials so far. A brief review is made of recent target-based and phenotypic studies based on the search for molecules with anti-Trypanosoma cruzi action. It also discusses how proteochemometric (PCM) modeling and microcrystal electron diffraction (MicroED) can help in the case of the lack of a 3D protein structure; more specifically, Trypanosoma cruzi carbonic anhydrase.
Collapse
Affiliation(s)
- Felipe Raposo Passos Mansoldo
- BIOINOVAR-Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (F.R.P.M.); (V.d.S.C.)
| | - Fabrizio Carta
- Neurofarba Department, Università degli Studi di Firenze, Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy; (F.C.); (A.A.)
| | - Andrea Angeli
- Neurofarba Department, Università degli Studi di Firenze, Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy; (F.C.); (A.A.)
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania
| | - Veronica da Silva Cardoso
- BIOINOVAR-Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (F.R.P.M.); (V.d.S.C.)
| | - Claudiu T. Supuran
- Neurofarba Department, Università degli Studi di Firenze, Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy; (F.C.); (A.A.)
| | - Alane Beatriz Vermelho
- BIOINOVAR-Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (F.R.P.M.); (V.d.S.C.)
| |
Collapse
|
40
|
Martín-Escolano J, Medina-Carmona E, Martín-Escolano R. Chagas Disease: Current View of an Ancient and Global Chemotherapy Challenge. ACS Infect Dis 2020; 6:2830-2843. [PMID: 33034192 DOI: 10.1021/acsinfecdis.0c00353] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chagas disease is a neglected tropical disease and a global public health issue. In terms of treatment, no progress has been made since the 1960s, when benznidazole and nifurtimox, two obsolete drugs still prescribed, were used to treat this disease. Hence, currently, there are no effective treatments available to tackle Chagas disease. Over the past 20 years, there has been an increasing interest in the disease. However, parasite genetic diversity, drug resistance, tropism, and complex life cycle, along with the limited understanding of the disease and inadequate methodologies and strategies, have resulted in the absence of new insights in drugs development and disappointing outcomes in clinical trials so far. In summary, new drugs are urgently needed. This Review considers the relevant aspects related to the lack of drugs for Chagas disease, resumes the advances in tools for drug discovery, and discusses the main features to be taken into account to develop new effective drugs.
Collapse
Affiliation(s)
- Javier Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | | | - Rubén Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| |
Collapse
|
41
|
Van den Kerkhof M, Sterckx YGJ, Leprohon P, Maes L, Caljon G. Experimental Strategies to Explore Drug Action and Resistance in Kinetoplastid Parasites. Microorganisms 2020; 8:E950. [PMID: 32599761 PMCID: PMC7356981 DOI: 10.3390/microorganisms8060950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022] Open
Abstract
Kinetoplastids are the causative agents of leishmaniasis, human African trypanosomiasis, and American trypanosomiasis. They are responsible for high mortality and morbidity in (sub)tropical regions. Adequate treatment options are limited and have several drawbacks, such as toxicity, need for parenteral administration, and occurrence of treatment failure and drug resistance. Therefore, there is an urgency for the development of new drugs. Phenotypic screening already allowed the identification of promising new chemical entities with anti-kinetoplastid activity potential, but knowledge on their mode-of-action (MoA) is lacking due to the generally applied whole-cell based approach. However, identification of the drug target is essential to steer further drug discovery and development. Multiple complementary techniques have indeed been used for MoA elucidation. In this review, the different 'omics' approaches employed to define the MoA or mode-of-resistance of current reference drugs and some new anti-kinetoplastid compounds are discussed.
Collapse
Affiliation(s)
- Magali Van den Kerkhof
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, 2610 Wilrijk, Belgium; (M.V.d.K.); (L.M.)
| | - Yann G.-J. Sterckx
- Laboratory of Medical Biochemistry (LMB), University of Antwerp, 2610 Wilrijk, Belgium;
| | - Philippe Leprohon
- Centre de Recherche en Infectiologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, 2610 Wilrijk, Belgium; (M.V.d.K.); (L.M.)
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, 2610 Wilrijk, Belgium; (M.V.d.K.); (L.M.)
| |
Collapse
|
42
|
Kempf DJ, Marsh KC. Assembling Pharma Resources to Tackle Diseases of Underserved Populations. ACS Med Chem Lett 2020; 11:1094-1100. [PMID: 32550987 DOI: 10.1021/acsmedchemlett.0c00051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/27/2020] [Indexed: 01/11/2023] Open
Abstract
Tropical diseases that disproportionally affect the world's poorest people have traditionally been neglected from research efforts toward the discovery and development of new and effective therapies. Over the past two decades, major global health funders have made efforts to bring together various research institutions to work together in these disease areas offering little or no commercial return. This work describes the genesis and growth of an informal program devoted to contributing to new therapies for neglected tropical diseases within the environment of a major biopharmaceutical company (AbbVie).
Collapse
|
43
|
Antileishmanial activity evaluation of thiazolidine-2,4-dione against Leishmania infantum and Leishmania braziliensis. Parasitol Res 2020; 119:2263-2274. [PMID: 32462293 DOI: 10.1007/s00436-020-06706-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/05/2020] [Indexed: 01/04/2023]
Abstract
Leishmaniasis is responsible for approximately 65,000 annual deaths. Despite the mortality data, drugs available for the treatment of patients are insufficient and have moderate therapeutic efficacy in addition to serious adverse effects, which makes the development of new drugs urgent. To achieve this goal, the integration of kinetic and DSF assays against parasitic validated targets, along with phenotypic assays, can help the identification and optimization of bioactive compounds. Pteridine reductase 1 (PTR1), a validated target in Leishmania sp., is responsible for the reduction of folate and biopterin to tetrahydrofolate and tetrahydrobiopterin, respectively, both of which are essential for cell growth. In addition to the in vitro evaluation of 16 thiazolidine-2,4-dione derivatives against Leishmania major PTR1 (LmPTR1), using the differential scanning fluorimetry (ThermoFluor®), phenotypic assays were employed to evaluate the compound effect over Leishmania braziliensis (MHOM/BR/75/M2903) and Leishmania infantum (MHOM/BR/74/PP75) promastigotes viability. The ThermoFluor® results show that thiazolidine-2,4-dione derivatives have micromolar affinity to the target and equivalent activity on Leishmania cells. 2b is the most potent compound against L. infantum (EC50 = 23.45 ± 4.54 μM), whereas 2a is the most potent against L. braziliensis (EC50 = 44.16 ± 5.77 μM). This result suggests that lipophilic substituents on either-meta and/or-para positions of the benzylidene ring increase the potency against L. infantum. On the other hand, compound 2c (CE50 = 49.22 ± 7.71 μM) presented the highest selectivity index.
Collapse
|
44
|
Pereira CA, Sayé M, Reigada C, Silber AM, Labadie GR, Miranda MR, Valera-Vera E. Computational approaches for drug discovery against trypanosomatid-caused diseases. Parasitology 2020; 147:611-633. [PMID: 32046803 PMCID: PMC10317681 DOI: 10.1017/s0031182020000207] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
During three decades, only about 20 new drugs have been developed for malaria, tuberculosis and all neglected tropical diseases (NTDs). This critical situation was reached because NTDs represent only 10% of health research investments; however, they comprise about 90% of the global disease burden. Computational simulations applied in virtual screening (VS) strategies are very efficient tools to identify pharmacologically active compounds or new indications for drugs already administered for other diseases. One of the advantages of this approach is the low time-consuming and low-budget first stage, which filters for testing experimentally a group of candidate compounds with high chances of binding to the target and present trypanocidal activity. In this work, we review the most common VS strategies that have been used for the identification of new drugs with special emphasis on those applied to trypanosomiasis and leishmaniasis. Computational simulations based on the selected protein targets or their ligands are explained, including the method selection criteria, examples of successful VS campaigns applied to NTDs, a list of validated molecular targets for drug development and repositioned drugs for trypanosomatid-caused diseases. Thereby, here we present the state-of-the-art of VS and drug repurposing to conclude pointing out the future perspectives in the field.
Collapse
Affiliation(s)
- Claudio A. Pereira
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Melisa Sayé
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Chantal Reigada
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Ariel M. Silber
- Laboratory of Biochemistry of Tryps – LaBTryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Guillermo R. Labadie
- Instituto de Química Rosario (IQUIR-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- Departamento de Química Orgánica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Mariana R. Miranda
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Edward Valera-Vera
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| |
Collapse
|
45
|
Álvarez-Bardón M, Pérez-Pertejo Y, Ordóñez C, Sepúlveda-Crespo D, Carballeira NM, Tekwani BL, Murugesan S, Martinez-Valladares M, García-Estrada C, Reguera RM, Balaña-Fouce R. Screening Marine Natural Products for New Drug Leads against Trypanosomatids and Malaria. Mar Drugs 2020; 18:E187. [PMID: 32244488 PMCID: PMC7230869 DOI: 10.3390/md18040187] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Neglected Tropical Diseases (NTD) represent a serious threat to humans, especially for those living in poor or developing countries. Almost one-sixth of the world population is at risk of suffering from these diseases and many thousands die because of NTDs, to which we should add the sanitary, labor and social issues that hinder the economic development of these countries. Protozoan-borne diseases are responsible for more than one million deaths every year. Visceral leishmaniasis, Chagas disease or sleeping sickness are among the most lethal NTDs. Despite not being considered an NTD by the World Health Organization (WHO), malaria must be added to this sinister group. Malaria, caused by the apicomplexan parasite Plasmodium falciparum, is responsible for thousands of deaths each year. The treatment of this disease has been losing effectiveness year after year. Many of the medicines currently in use are obsolete due to their gradual loss of efficacy, their intrinsic toxicity and the emergence of drug resistance or a lack of adherence to treatment. Therefore, there is an urgent and global need for new drugs. Despite this, the scant interest shown by most of the stakeholders involved in the pharmaceutical industry makes our present therapeutic arsenal scarce, and until recently, the search for new drugs has not been seriously addressed. The sources of new drugs for these and other pathologies include natural products, synthetic molecules or repurposing drugs. The most frequent sources of natural products are microorganisms, e.g., bacteria, fungi, yeasts, algae and plants, which are able to synthesize many drugs that are currently in use (e.g. antimicrobials, antitumor, immunosuppressants, etc.). The marine environment is another well-established source of bioactive natural products, with recent applications against parasites, bacteria and other pathogens which affect humans and animals. Drug discovery techniques have rapidly advanced since the beginning of the millennium. The combination of novel techniques that include the genetic modification of pathogens, bioimaging and robotics has given rise to the standardization of High-Performance Screening platforms in the discovery of drugs. These advancements have accelerated the discovery of new chemical entities with antiparasitic effects. This review presents critical updates regarding the use of High-Throughput Screening (HTS) in the discovery of drugs for NTDs transmitted by protozoa, including malaria, and its application in the discovery of new drugs of marine origin.
Collapse
Affiliation(s)
- María Álvarez-Bardón
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Yolanda Pérez-Pertejo
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - César Ordóñez
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Daniel Sepúlveda-Crespo
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Nestor M. Carballeira
- Department of Chemistry, University of Puerto Rico, Río Piedras 00925-2537, San Juan, Puerto Rico;
| | - Babu L. Tekwani
- Department of Infectious Diseases, Division of Drug Discovery, Southern Research, Birmingham, AL 35205, USA;
| | - Sankaranarayanan Murugesan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani 333031, India;
| | - Maria Martinez-Valladares
- Department of Animal Health, Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, 24346 León, Spain;
| | - Carlos García-Estrada
- INBIOTEC (Instituto de Biotecnología de León), Avda. Real 1-Parque Científico de León, 24006 León, Spain;
| | - Rosa M. Reguera
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Rafael Balaña-Fouce
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| |
Collapse
|
46
|
Wall RJ, Carvalho S, Milne R, Bueren-Calabuig JA, Moniz S, Cantizani-Perez J, MacLean L, Kessler A, Cotillo I, Sastry L, Manthri S, Patterson S, Zuccotto F, Thompson S, Martin J, Marco M, Miles TJ, De Rycker M, Thomas MG, Fairlamb AH, Gilbert IH, Wyllie S. The Q i Site of Cytochrome b is a Promiscuous Drug Target in Trypanosoma cruzi and Leishmania donovani. ACS Infect Dis 2020; 6:515-528. [PMID: 31967783 PMCID: PMC7076694 DOI: 10.1021/acsinfecdis.9b00426] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Indexed: 01/29/2023]
Abstract
Available treatments for Chagas' disease and visceral leishmaniasis are inadequate, and there is a pressing need for new therapeutics. Drug discovery efforts for both diseases principally rely upon phenotypic screening. However, the optimization of phenotypically active compounds is hindered by a lack of information regarding their molecular target(s). To combat this issue we initiate target deconvolution studies at an early stage. Here, we describe comprehensive genetic and biochemical studies to determine the targets of three unrelated phenotypically active compounds. All three structurally diverse compounds target the Qi active-site of cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Our studies go on to identify the Qi site as a promiscuous drug target in Leishmania donovani and Trypanosoma cruzi with a propensity to rapidly mutate. Strategies to rapidly identify compounds acting via this mechanism are discussed to ensure that drug discovery portfolios are not overwhelmed with inhibitors of a single target.
Collapse
Affiliation(s)
- Richard J. Wall
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Sandra Carvalho
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Rachel Milne
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Juan A. Bueren-Calabuig
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sonia Moniz
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | | | - Lorna MacLean
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Albane Kessler
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Ignacio Cotillo
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Lalitha Sastry
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sujatha Manthri
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Stephen Patterson
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Fabio Zuccotto
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Stephen Thompson
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Julio Martin
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Maria Marco
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | | | - Manu De Rycker
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Michael G. Thomas
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Alan H. Fairlamb
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Ian H. Gilbert
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Susan Wyllie
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| |
Collapse
|
47
|
Eyssen LEA, Coetzer TH. Validation of ligands targeting metacaspase-2 (MCA2) from Trypanosoma brucei brucei and their application to MCA5 from T. congolense as possible trypanocides. J Mol Graph Model 2020; 97:107579. [PMID: 32197135 DOI: 10.1016/j.jmgm.2020.107579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 11/29/2022]
Abstract
Metacaspases (MCAs) are ideal drug and diagnostic targets for animal and human African trypanosomiasis, as these cysteine peptidases are absent from the metazoan kingdom and have been implicated in the parasite cell cycle and cell death. Tsetse fly-transmitted trypanosomes that live free in the bloodstream and/or cerebrospinal fluid of the mammalian host cause animal and human African trypanosomiasis (nagana or sleeping sickness respectively). Chemotherapy and chemoprophylaxis are the main forms of control, but in contrast to human trypanocides, the veterinary drugs are old and drug resistance is on the increase. A peptidomimetic library targeting the MCA2 from Trypanosoma brucei brucei has ligands with low IC50 values, some of which were antiparasitic. This study validates the inhibitory activity of these ligands using the protein structure solved by X-ray diffraction after the ligand library was published. Water molecules were shown to be important in substrate binding and strategies to improve the efficacy of these ligands are highlighted. These ligands appear to be pan-specific as they were docked into the active site of the homology modelled MCA5 of animal infective Trypanosoma congolense with similar binding energies and conformations.
Collapse
Affiliation(s)
- L E-A Eyssen
- Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville, 3209, South Africa
| | - Theresa Ht Coetzer
- Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Pietermaritzburg Campus), Private Bag X01, Scottsville, 3209, South Africa.
| |
Collapse
|
48
|
Sebastian-Perez V, García-Rubia A, Seif El-Din SH, Sabra ANA, El-Lakkany NM, William S, Blundell TL, Maes L, Martinez A, Campillo NE, Botros SS, Gil C. Deciphering the enzymatic target of a new family of antischistosomal agents bearing a quinazoline scaffold using complementary computational tools. J Enzyme Inhib Med Chem 2020; 35:511-523. [PMID: 31939312 PMCID: PMC7717570 DOI: 10.1080/14756366.2020.1712595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A previous phenotypic screening campaign led to the identification of a quinazoline derivative with promising in vitro activity against Schistosoma mansoni. Follow-up studies of the antischistosomal potential of this candidate are presented here. The in vivo studies in a S. mansoni mouse model show a significant reduction of total worms and a complete disappearance of immature eggs when administered concomitantly with praziquantel in comparison with the administration of praziquantel alone. This fact is of utmost importance because eggs are responsible for the pathology and transmission of the disease. Subsequently, the chemical optimisation of the structure in order to improve the metabolic stability of the parent compound was carried out leading to derivatives with improved drug-like properties. Additionally, the putative target of this new class of antischistosomal compounds was envisaged by using computational tools and the binding mode to the target enzyme, aldose reductase, was proposed.
Collapse
Affiliation(s)
| | | | | | | | | | - Samia William
- Parasitology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Ana Martinez
- Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | | | - Sanaa S Botros
- Pharmacology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Carmen Gil
- Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
49
|
Linciano P, Cullia G, Borsari C, Santucci M, Ferrari S, Witt G, Gul S, Kuzikov M, Ellinger B, Santarém N, Cordeiro da Silva A, Conti P, Bolognesi ML, Roberti M, Prati F, Bartoccini F, Retini M, Piersanti G, Cavalli A, Goldoni L, Bertozzi SM, Bertozzi F, Brambilla E, Rizzo V, Piomelli D, Pinto A, Bandiera T, Costi MP. Identification of a 2,4-diaminopyrimidine scaffold targeting Trypanosoma brucei pteridine reductase 1 from the LIBRA compound library screening campaign. Eur J Med Chem 2020; 189:112047. [PMID: 31982652 DOI: 10.1016/j.ejmech.2020.112047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Abstract
The LIBRA compound library is a collection of 522 non-commercial molecules contributed by various Italian academic laboratories. These compounds have been designed and synthesized during different medicinal chemistry programs and are hosted by the Italian Institute of Technology. We report the screening of the LIBRA compound library against Trypanosoma brucei and Leishmania major pteridine reductase 1, TbPTR1 and LmPTR1. Nine compounds were active against parasitic PTR1 and were selected for cell-based parasite screening, as single agents and in combination with methotrexate (MTX). The most interesting TbPTR1 inhibitor identified was 4-(benzyloxy)pyrimidine-2,6-diamine (LIB_66). Subsequently, six new LIB_66 derivatives were synthesized to explore its Structure-Activity-Relationship (SAR) and absorption, distribution, metabolism, excretion and toxicity (ADMET) properties. The results indicate that PTR1 has a preference to bind inhibitors, which resemble its biopterin/folic acid substrates, such as the 2,4-diaminopyrimidine derivatives.
Collapse
Affiliation(s)
- Pasquale Linciano
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Gregorio Cullia
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Chiara Borsari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Matteo Santucci
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Stefania Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Nuno Santarém
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal and Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, 4150-180, Porto, Portugal
| | - Anabela Cordeiro da Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal and Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, 4150-180, Porto, Portugal; Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Marinella Roberti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Federica Prati
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Francesca Bartoccini
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, Section of Chemistry, University of Urbino "Carlo Bo", Piazza Rinascimento 6, 61029, Urbino, Italy
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, Section of Chemistry, University of Urbino "Carlo Bo", Piazza Rinascimento 6, 61029, Urbino, Italy
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy; Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Luca Goldoni
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Sine Mandrup Bertozzi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Fabio Bertozzi
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Enzo Brambilla
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Vincenzo Rizzo
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, 92697-4625, USA
| | - Andrea Pinto
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133, Milan, Italy
| | - Tiziano Bandiera
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy.
| |
Collapse
|
50
|
Kwofie SK, Broni E, Dankwa B, Enninful KS, Kwarko GB, Darko L, Durvasula R, Kempaiah P, Rathi B, Miller Iii WA, Yaya A, Wilson MD. Outwitting an Old Neglected Nemesis: A Review on Leveraging Integrated Data-Driven Approaches to Aid in Unraveling of Leishmanicides of Therapeutic Potential. Curr Top Med Chem 2020; 20:349-366. [PMID: 31994465 DOI: 10.2174/1568026620666200128160454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/20/2019] [Accepted: 09/12/2019] [Indexed: 11/22/2022]
Abstract
The global prevalence of leishmaniasis has increased with skyrocketed mortality in the past decade. The causative agent of leishmaniasis is Leishmania species, which infects populations in almost all the continents. Prevailing treatment regimens are consistently inefficient with reported side effects, toxicity and drug resistance. This review complements existing ones by discussing the current state of treatment options, therapeutic bottlenecks including chemoresistance and toxicity, as well as drug targets. It further highlights innovative applications of nanotherapeutics-based formulations, inhibitory potential of leishmanicides, anti-microbial peptides and organometallic compounds on leishmanial species. Moreover, it provides essential insights into recent machine learning-based models that have been used to predict novel leishmanicides and also discusses other new models that could be adopted to develop fast, efficient, robust and novel algorithms to aid in unraveling the next generation of anti-leishmanial drugs. A plethora of enriched functional genomic, proteomic, structural biology, high throughput bioassay and drug-related datasets are currently warehoused in both general and leishmania-specific databases. The warehoused datasets are essential inputs for training and testing algorithms to augment the prediction of biotherapeutic entities. In addition, we demonstrate how pharmacoinformatics techniques including ligand-, structure- and pharmacophore-based virtual screening approaches have been utilized to screen ligand libraries against both modeled and experimentally solved 3D structures of essential drug targets. In the era of data-driven decision-making, we believe that highlighting intricately linked topical issues relevant to leishmanial drug discovery offers a one-stop-shop opportunity to decipher critical literature with the potential to unlock implicit breakthroughs.
Collapse
Affiliation(s)
- Samuel K Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra, Ghana.,West African Center for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States
| | - Emmanuel Broni
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra, Ghana
| | - Bismark Dankwa
- Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon, Accra, Ghana
| | - Kweku S Enninful
- Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon, Accra, Ghana
| | - Gabriel B Kwarko
- West African Center for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Louis Darko
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra, Ghana
| | - Ravi Durvasula
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States
| | - Prakasha Kempaiah
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States
| | - Brijesh Rathi
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States.,Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi, 110007, India
| | - Whelton A Miller Iii
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States.,Department of Chemistry, Physics, & Engineering, Lincoln University, Lincoln University, PA 19352, United States.,Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Abu Yaya
- Department of Materials Science and Engineering, College of Basic & Applied Sciences, University of Ghana, Legon, Ghana
| | - Michael D Wilson
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States.,Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon, Accra, Ghana
| |
Collapse
|