1
|
Sulik M, Fontinha D, Steverding D, Sobczak S, Antoszczak M, Prudêncio M, Huczyński A. Unexpected rearrangement of ivermectin in the synthesis of new derivatives with trypanocidal and antiplasmodial activities. Eur J Med Chem 2024; 263:115951. [PMID: 37988797 DOI: 10.1016/j.ejmech.2023.115951] [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: 10/06/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Ivermectin is a sixteen-membered macrolactone "wonder drug" of Nobel prize-honored distinction that exhibits a wide range of antiparasitic activities. It has been used for almost four decades in the treatment of various parasitic diseases in humans and animals. In this paper, we describe the synthesis of the first-in-class ivermectin derivatives obtained via derivatization of the C13 position, along with the unexpected rearrangement of the oxahydrindene (hexahydrobenzofuran) unit of the macrolide ring. The structural investigation of the rearrangement has been performed using the single-crystal X-ray diffraction method. The antiparasitic and cytotoxic activities of the newly synthesized derivatives were determined in vitro with the bloodstream form of Trypanosoma brucei brucei, the hepatic stage of Plasmodium berghei, and human leukemia HL-60 cells. The compounds with the highest trypanocidal activity were the C13-epi-2-chloroacetamide analogs of native (6h) or rearranged (7h) ivermectin. Both 6h and 7h displayed trypanocidal activities within a similar mid-nanomolar concentration range as the commercially used trypanocides suramin and ethidium bromide. Furthermore, 6h and 7h exhibited a comparable cytotoxic to trypanocidal ratio as the reference drug ethidium bromide. The double-modified compound 7a (C13-epi-acetamide of rearranged ivermectin) exhibited the highest activity against P. berghei grown in human hepatoma cells, which was 2.5 times higher than that of ivermectin. The findings of this study suggest that C13-epi-amide derivatives of ivermectin are suitable leads in the rational development of new antiparasitic agents.
Collapse
Affiliation(s)
- Michał Sulik
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614 Poznań, Poland
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Dietmar Steverding
- Bob Champion Research & Education Building, Norwich Medical School, University of East Anglia, Norwich, UK
| | - Szymon Sobczak
- Department of Materials Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614 Poznań, Poland
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614 Poznań, Poland
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614 Poznań, Poland.
| |
Collapse
|
2
|
Sulik M, Antoszczak M, Huczyński A, Steverding D. Antiparasitic activity of ivermectin: Four decades of research into a "wonder drug". Eur J Med Chem 2023; 261:115838. [PMID: 37793327 DOI: 10.1016/j.ejmech.2023.115838] [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: 08/22/2023] [Revised: 09/17/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
Parasitic diseases still pose a serious threat to human and animal health, particularly for millions of people and their livelihoods in low-income countries. Therefore, research into the development of effective antiparasitic drugs remains a priority. Ivermectin, a sixteen-membered macrocyclic lactone, exhibits a broad spectrum of antiparasitic activities, which, combined with its low toxicity, has allowed the drug to be widely used in the treatment of parasitic diseases affecting humans and animals. In addition to its licensed use against river blindness and strongyloidiasis in humans, and against roundworm and arthropod infestations in animals, ivermectin is also used "off-label" to treat many other worm-related parasitic diseases, particularly in domestic animals. In addition, several experimental studies indicate that ivermectin displays also potent activity against viruses, bacteria, protozoans, trematodes, and insects. This review article summarizes the last 40 years of research on the antiparasitic effects of ivermectin, and the use of the drug in the treatment of parasitic diseases in humans and animals.
Collapse
Affiliation(s)
- Michał Sulik
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614, Poznań, Poland
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614, Poznań, Poland
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61‒614, Poznań, Poland.
| | - Dietmar Steverding
- Bob Champion Research & Education Building, Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
3
|
Fonte M, Fontinha D, Moita D, Caño-Prades O, Avalos-Padilla Y, Fernàndez-Busquets X, Prudêncio M, Gomes P, Teixeira C. New 4-(N-cinnamoylbutyl)aminoacridines as potential multi-stage antiplasmodial leads. Eur J Med Chem 2023; 258:115575. [PMID: 37390511 DOI: 10.1016/j.ejmech.2023.115575] [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: 04/14/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
A novel family of 4-aminoacridine derivatives was obtained by linking this heteroaromatic core to different trans-cinnamic acids. The 4-(N-cinnamoylbutyl)aminoacridines obtained exhibited in vitro activity in the low- or sub-micromolar range against (i) hepatic stages of Plasmodium berghei, (ii) erythrocytic forms of Plasmodium falciparum, and (iii) early and mature gametocytes of Plasmodium falciparum. The most active compound, having a meta-fluorocinnamoyl group linked to the acridine core, was 20- and 120-fold more potent, respectively, against the hepatic and gametocyte stages of Plasmodium infection than the reference drug, primaquine. Moreover, no cytotoxicity towards mammalian and red blood cells at the concentrations tested was observed for any of the compounds under investigation. These novel conjugates represent promising leads for the development of new multi-target antiplasmodials.
Collapse
Affiliation(s)
- Mélanie Fonte
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal.
| | - Diana Fontinha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Diana Moita
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Omar Caño-Prades
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Yunuen Avalos-Padilla
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Xavier Fernàndez-Busquets
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Spain
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal.
| | - Cátia Teixeira
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Gyros Protein Technologies Inc., Tucson, AZ, USA
| |
Collapse
|
4
|
Breiterová KH, Ritomská A, Fontinha D, Křoustková J, Suchánková D, Hošťálková A, Šafratová M, Kohelová E, Peřinová R, Vrabec R, Francisco D, Prudêncio M, Cahlíková L. Derivatives of Amaryllidaceae Alkaloid Ambelline as Selective Inhibitors of Hepatic Stage of Plasmodium berghei Infection In Vitro. Pharmaceutics 2023; 15:pharmaceutics15031007. [PMID: 36986868 PMCID: PMC10056443 DOI: 10.3390/pharmaceutics15031007] [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: 12/27/2022] [Revised: 02/28/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
The incidence rate of malaria and the ensuing mortality prompts the development of novel antimalarial drugs. In this work, the activity of twenty-eight Amaryllidaceae alkaloids (1-28) belonging to seven different structural types was assessed, as well as twenty semisynthetic derivatives of the β-crinane alkaloid ambelline (28a-28t) and eleven derivatives of the α-crinane alkaloid haemanthamine (29a-29k) against the hepatic stage of Plasmodium infection. Six of these derivatives (28h, 28m, 28n and 28r-28t) were newly synthesized and structurally identified. The most active compounds, 11-O-(3,5-dimethoxybenzoyl)ambelline (28m) and 11-O-(3,4,5-trimethoxybenzoyl)ambelline (28n), displayed IC50 values in the nanomolar range of 48 and 47 nM, respectively. Strikingly, the derivatives of haemanthamine (29) with analogous substituents did not display any significant activity, even though their structures are quite similar. Interestingly, all active derivatives were strictly selective against the hepatic stage of infection, as they did not demonstrate any activity against the blood stage of Plasmodium infection. As the hepatic stage is a bottleneck of the plasmodial infection, liver-selective compounds can be considered crucial for further development of the malaria prophylactics.
Collapse
Affiliation(s)
- Kateřina Hradiská Breiterová
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Aneta Ritomská
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Diana Fontinha
- Prudêncio Lab, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Edf. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Jana Křoustková
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Daniela Suchánková
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Anna Hošťálková
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Marcela Šafratová
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Eliška Kohelová
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Rozálie Peřinová
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Rudolf Vrabec
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Denise Francisco
- Prudêncio Lab, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Edf. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Miguel Prudêncio
- Prudêncio Lab, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Edf. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Lucie Cahlíková
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| |
Collapse
|
5
|
Muema JM, Bargul JL, Obonyo MA, Njeru SN, Matoke-Muhia D, Mutunga JM. Contemporary exploitation of natural products for arthropod-borne pathogen transmission-blocking interventions. Parasit Vectors 2022; 15:298. [PMID: 36002857 PMCID: PMC9404607 DOI: 10.1186/s13071-022-05367-8] [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: 02/24/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
An integrated approach to innovatively counter the transmission of various arthropod-borne diseases to humans would benefit from strategies that sustainably limit onward passage of infective life cycle stages of pathogens and parasites to the insect vectors and vice versa. Aiming to accelerate the impetus towards a disease-free world amid the challenges posed by climate change, discovery, mindful exploitation and integration of active natural products in design of pathogen transmission-blocking interventions is of high priority. Herein, we provide a review of natural compounds endowed with blockade potential against transmissible forms of human pathogens reported in the last 2 decades from 2000 to 2021. Finally, we propose various translational strategies that can exploit these pathogen transmission-blocking natural products into design of novel and sustainable disease control interventions. In summary, tapping these compounds will potentially aid in integrated combat mission to reduce disease transmission trends.
Collapse
Affiliation(s)
- Jackson M Muema
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.,International Centre of Insect Physiology and Ecology (Icipe), P.O. Box 30772, Nairobi, 00100, Kenya
| | - Meshack A Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Egerton, 20115, Kenya
| | - Sospeter N Njeru
- Centre for Traditional Medicine and Drug Research (CTMDR), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - Damaris Matoke-Muhia
- Centre for Biotechnology Research Development (CBRD), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - James M Mutunga
- Department of Biological Sciences, Mount Kenya University (MKU), P.O. Box 54, Thika, 01000, Kenya.,School of Engineering Design, Technology and Professional Programs, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
6
|
Chemical and Pharmacological Properties of Decoquinate: A Review of Its Pharmaceutical Potential and Future Perspectives. Pharmaceutics 2022; 14:pharmaceutics14071383. [PMID: 35890280 PMCID: PMC9315532 DOI: 10.3390/pharmaceutics14071383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022] Open
Abstract
Decoquinate (DQ) is an antimicrobial agent commonly used as a feed additive for birds for human consumption. Its use as an additive is well established, but DQ has the potential for therapy as an antimicrobial drug for veterinary treatment and its optimized derivatives and/or formulations, mainly nanoformulations, have antimicrobial activity against pathogens that infect humans. However, DQ has a high partition coefficient and low solubility in aqueous fluids, and these biopharmaceutical properties have limited its use in humans. In this review, we highlight the antimicrobial activity and pharmacokinetic properties of DQ and highlight the solutions currently under investigation to overcome these drawbacks. A literature search was conducted focusing on the use of decoquinate against various infectious diseases in humans and animals. The search was conducted in several databases, including scientific and patent databases. Pharmaceutical nanotechnology and medicinal chemistry are the tools of choice to achieve human applications, and most of these applications have been able to improve the biopharmaceutical properties and pharmacokinetic profile of DQ. Based on the results presented here, DQ prototypes could be tested in clinical trials for human application in the coming years.
Collapse
|
7
|
Khandelwal A, Arez F, Alves PM, Badolo L, Brito C, Fischli C, Fontinha D, Oeuvray C, Prudêncio M, Rottmann M, Wilkins J, Yalkinoglu Ö, Bagchus WM, Spangenberg T. Translation of liver stage activity of M5717, a Plasmodium elongation factor 2 inhibitor: from bench to bedside. Malar J 2022; 21:151. [PMID: 35570264 PMCID: PMC9107587 DOI: 10.1186/s12936-022-04171-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
Background Targeting the asymptomatic liver stage of Plasmodium infection through chemoprevention could become a key intervention to reduce malaria-associated incidence and mortality. Methods M5717, a Plasmodium elongation factor 2 inhibitor, was assessed in vitro and in vivo with readily accessible Plasmodium berghei parasites. In an animal refinement, reduction, replacement approach, the in vitro IC99 value was used to feed a Population Pharmacokinetics modelling and simulation approach to determine meaningful effective doses for a subsequent Plasmodium sporozoite-induced volunteer infection study. Results Doses of 100 and 200 mg would provide exposures exceeding IC99 in 96 and 100% of the simulated population, respectively. Conclusions This approach has the potential to accelerate the search for new anti-malarials, to reduce the number of healthy volunteers needed in a clinical study and decrease and refine the animal use in the preclinical phase. Graphical Abstract ![]()
Collapse
|
8
|
Dong Y, Sonawane Y, Maher SP, Zeeman AM, Chaumeau V, Vantaux A, Cooper CA, Chiu FCK, Ryan E, McLaren J, Chen G, Wittlin S, Witkowski B, Nosten F, Sriraghavan K, Kyle DE, Kocken CHM, Charman SA, Vennerstrom JL. Metabolic, Pharmacokinetic, and Activity Profile of the Liver Stage Antimalarial (RC-12). ACS OMEGA 2022; 7:12401-12411. [PMID: 35449901 PMCID: PMC9016807 DOI: 10.1021/acsomega.2c01099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/18/2022] [Indexed: 06/14/2023]
Abstract
The catechol derivative RC-12 (WR 27653) (1) is one of the few non-8-aminoquinolines with good activity against hypnozoites in the gold-standard Plasmodium cynomolgi-rhesus monkey (Macaca mulatta) model, but in a small clinical trial, it had no efficacy against Plasmodium vivax hypnozoites. In an attempt to better understand the pharmacokinetic and pharmacodynamic profile of 1 and to identify potential active metabolites, we now describe the phase I metabolism, rat pharmacokinetics, and in vitro liver-stage activity of 1 and its metabolites. Compound 1 had a distinct metabolic profile in human vs monkey liver microsomes, and the data suggested that the O-desmethyl, combined O-desmethyl/N-desethyl, and N,N-didesethyl metabolites (or a combination thereof) could potentially account for the superior liver stage antimalarial efficacy of 1 in rhesus monkeys vs that seen in humans. Indeed, the rate of metabolism was considerably lower in human liver microsomes in comparison to rhesus monkey microsomes, as was the formation of the combined O-desmethyl/N-desethyl metabolite, which was the only metabolite tested that had any activity against liver-stage P. vivax; however, it was not consistently active against liver-stage P. cynomolgi. As 1 and all but one of its identified Phase I metabolites had no in vitro activity against P. vivax or P. cynomolgi liver-stage malaria parasites, we suggest that there may be additional unidentified active metabolites of 1 or that the exposure of 1 achieved in the reported unsuccessful clinical trial of this drug candidate was insufficient to kill the P. vivax hypnozoites.
Collapse
Affiliation(s)
- Yuxiang Dong
- College
of Pharmacy, University of Nebraska Medical
Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yogesh Sonawane
- College
of Pharmacy, University of Nebraska Medical
Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Steven P. Maher
- Center
for Tropical and Emerging Global Diseases, University of Georgia, 370 Coverdell
Center, 500 D.W. Brooks Drive, Athens, Georgia 30602, United States
| | - Anne-Marie Zeeman
- Department
of Parasitology, Biomedical Primate Research
Centre, P.O. Box 3306, 2280 GH Rijswijk, The Netherlands
| | - Victor Chaumeau
- Shoklo
Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit,
Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Road, Mae Sot, Tak 63110, Thailand
- Centre for
Tropical Medicine and Global Health, Nuffield Department of Medicine
Research building, University of Oxford
Old Road Campus, Oxford OX3 7DQ, U.K.
| | - Amélie Vantaux
- Malaria
Molecular Epidemiology Unit, Institut Pasteur
du Cambodge, 5 Boulevard Monivong, P.O. Box 983, Phnom
Penh 120 210, Cambodia
| | - Caitlin A. Cooper
- Center
for Tropical and Emerging Global Diseases, University of Georgia, 370 Coverdell
Center, 500 D.W. Brooks Drive, Athens, Georgia 30602, United States
| | - Francis C. K. Chiu
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Eileen Ryan
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jenna McLaren
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Gong Chen
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Sergio Wittlin
- Department
of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland
| | - Benoît Witkowski
- Malaria
Molecular Epidemiology Unit, Institut Pasteur
du Cambodge, 5 Boulevard Monivong, P.O. Box 983, Phnom
Penh 120 210, Cambodia
| | - François Nosten
- Shoklo
Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit,
Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Road, Mae Sot, Tak 63110, Thailand
- Centre for
Tropical Medicine and Global Health, Nuffield Department of Medicine
Research building, University of Oxford
Old Road Campus, Oxford OX3 7DQ, U.K.
| | - Kamaraj Sriraghavan
- College
of Pharmacy, University of Nebraska Medical
Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Dennis E. Kyle
- Center
for Tropical and Emerging Global Diseases, University of Georgia, 370 Coverdell
Center, 500 D.W. Brooks Drive, Athens, Georgia 30602, United States
| | - Clemens H. M. Kocken
- Department
of Parasitology, Biomedical Primate Research
Centre, P.O. Box 3306, 2280 GH Rijswijk, The Netherlands
| | - Susan A. Charman
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jonathan L. Vennerstrom
- College
of Pharmacy, University of Nebraska Medical
Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
9
|
Lopes EA, Mestre R, Fontinha D, Legac J, Pei JV, Sanches-Vaz M, Mori M, Lehane AM, Rosenthal PJ, Prudêncio M, Santos MM. Discovery of spirooxadiazoline oxindoles with dual-stage antimalarial activity. Eur J Med Chem 2022; 236:114324. [DOI: 10.1016/j.ejmech.2022.114324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/21/2022]
|
10
|
Wang M, Tang T, Li R, Huang Z, Ling D, Zheng L, Ding Y, Liu T, Xu W, Zhu F, Min H, Boonhok R, Mao F, Zhu J, Li X, Jiang L, Li J. Drug Repurposing of Quisinostat to Discover Novel Plasmodium falciparum HDAC1 Inhibitors with Enhanced Triple-Stage Antimalarial Activity and Improved Safety. J Med Chem 2022; 65:4156-4181. [PMID: 35175762 DOI: 10.1021/acs.jmedchem.1c01993] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our previous work found that the clinical histone deacetylase (HDAC) inhibitor quisinostat exhibited a significant antimalarial effect but with severe toxicity. In this work, 35 novel derivatives were designed and synthesized based on quisinostat as the lead compound, and their in vitro antimalarial activities and cytotoxicities were systematically evaluated. Among them, JX35 showed potent inhibition against both wild-type and multidrug-resistant parasite strains and displayed a significant in vivo killing effect against all life cycles of parasites, including the blood stage, liver stage, and gametocyte stage, indicating its potential for the simultaneous treatment, chemoprevention, and blockage of malaria transmission. Compared with quisinostat, JX35 exhibited stronger antimalarial efficacy, more adequate safety, and good pharmacokinetic properties. Additionally, mechanistic studies via molecular docking studies, induced PfHDAC1/2 knockdown assays, and PfHDAC1 enzyme inhibition assays jointly indicated that the antimalarial target of JX35 was PfHDAC1. In summary, we discovered the promising candidate PfHDAC1 inhibitor JX35, which showed stronger triple-stage antimalarial effects and lower toxicity than quisinostat.
Collapse
Affiliation(s)
- Manjiong Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Tongke Tang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, P.R. China
| | - Ruoxi Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhenghui Huang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dazheng Ling
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lulu Zheng
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan Ding
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Feng Zhu
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Hui Min
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Rachasak Boonhok
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Fei Mao
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jin Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lubin Jiang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, P.R. China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.,College of Pharmacy and Chemistry, Dali University, 5 Xue Ren Road, Dali 671000, China.,Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| |
Collapse
|
11
|
Abid M, Singh S, Egan TJ, Joshi MC. Structural activity relationship of metallo-aminoquines as a next generation antimalarials. Curr Top Med Chem 2022; 22:436-472. [PMID: 34986771 DOI: 10.2174/1568026622666220105103751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 11/22/2022]
Abstract
Apicomplexian parasite of the genus Plasmodium is the causative agent of malaria, one of the most devastating, furious and common infectious disease throughout the world. According to the latest World malaria report, there were 229 million cases of malaria in 2019 majorly consisting of children under 5 years of age. Some of known analogues viz. quinine, quinoline-containing compounds have been used for last century in the clinical treatment of malaria. Past few decades have witnessed the emergence of multi-drug resistance (MDR) strains of Plasmodium species to existing antimalarials pressing the need for new drug candidates. For the past few decades bioorganometallic approach to malaria therapy has been introduced which led to the discovery of noval metalcontaining aminoquinolines analogues viz. ferroquine (FQ or 1), Ruthenoquine (RQ or 2) and other related potent metal-analogues. It observed that some metal containing analogues (Fe-, Rh-, Ru-, Re-, Au-, Zn-, Cr-, Pd-, Sn-, Cd-, Ir-, Co-, Cu-, and Mn-aminoquines) were more potent; however, some were equally potent as Chloroquine (CQ) and 1. This is probably due to the intertion of metals in the CQ via various approaches, which might be a very attractive strategy to develop a SAR of novel metal containing antimalarials. Thus, this review aims to summarize the SAR of metal containing aminoquines towards the discovery of potent antimalarial hybrids to provide an insight for rational designs of more effective and less toxic metal containing amoniquines.
Collapse
Affiliation(s)
- Mohammad Abid
- Department of Biosciences, Jamia Millia Islamia University, Jamia Nagar, New Delhi-110025, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Mehroli Road, New Delhi-110067, India
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town-7700, South Africa
| | - Mukesh C Joshi
- Dept. of Chemistry, Motilal Nehru College, University of Delhi, Benito Juarez marg, South Campus, New Delhi-110021. India
| |
Collapse
|
12
|
Upadhyay C, Sharma N, Kumar S, Sharma PP, Fontinha D, Chhikara BS, Mukherjee B, Kumar D, Prudencio M, Singh AP, Poonam. Synthesis of the new analogs of morpholine and their antiplasmodial evaluation against the human malaria parasite Plasmodium falciparum. NEW J CHEM 2022. [DOI: 10.1039/d1nj04198c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A series of morpholine analogs functionalized with hydroxyethylamine (HEA) pharmacophore was synthesized and assayed for the initial screening against Plasmodium falciparum 3D7 in culture, which suggested that analog 6k is a hit molecule with an inhibitory concentration of 5.059 ± 0.2036 μM.
Collapse
Affiliation(s)
- Charu Upadhyay
- Department of Chemistry, Miranda House, University of Delhi, Delhi, India
| | - Neha Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
| | - Sumit Kumar
- Department of Chemistry, Miranda House, University of Delhi, Delhi, India
| | - Prem Prakash Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
| | - Diana Fontinha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | | | - Budhaditya Mukherjee
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur-721302, India
| | - Dhruv Kumar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University Uttar Pradesh, Noida, India
| | - Miguel Prudencio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Agam P. Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi, Delhi, India
| |
Collapse
|
13
|
Jansongsaeng S, Srimongkolpithak N, Pengon J, Kamchonwongpaisan S, Khotavivattana T. 5-Phenoxy Primaquine Analogs and the Tetraoxane Hybrid as Antimalarial Agents. Molecules 2021; 26:molecules26133991. [PMID: 34208832 PMCID: PMC8272044 DOI: 10.3390/molecules26133991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 12/04/2022] Open
Abstract
The rapid emergence of drug resistance to the current antimalarial agents has led to the urgent need for the discovery of new and effective compounds. In this work, a series of 5-phenoxy primaquine analogs with 8-aminoquinoline core (7a–7h) was synthesized and investigated for their antimalarial activity against Plasmodium falciparum. Most analogs showed improved blood antimalarial activity compared to the original primaquine. To further explore a drug hybrid strategy, a conjugate compound between tetraoxane and the representative 5-phenoxy-primaquine analog 7a was synthesized. In our work, the hybrid compound 12 exhibited almost a 30-fold increase in the blood antimalarial activity (IC50 = 0.38 ± 0.11 μM) compared to that of primaquine, with relatively low toxicity against mammalian cells (SI = 45.61). Furthermore, we found that these 5-phenoxy primaquine analogs and the hybrid exhibit significant heme polymerization inhibition, an activity similar to that of chloroquine, which could contribute to their improved antimalarial activity. The 5-phenoxy primaquine analogs and the tetraoxane hybrid could serve as promising candidates for the further development of antimalarial agents.
Collapse
Affiliation(s)
- Somruedee Jansongsaeng
- Centre of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Nitipol Srimongkolpithak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand; (N.S.); (J.P.); (S.K.)
| | - Jutharat Pengon
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand; (N.S.); (J.P.); (S.K.)
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand; (N.S.); (J.P.); (S.K.)
| | - Tanatorn Khotavivattana
- Centre of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
- Correspondence: ; Tel.: +66-2-218-7621
| |
Collapse
|
14
|
Shim S, Park H, Mishra NK, Kim IS, Lee JK, Lee K, Jalani HB, Choi Y. Catalyst‐Free One‐Pot Multi‐Component Synthesis of 2‐Substituted Quinazolin‐4‐carboxamides from 2‐Aminophenyl‐2‐oxoacetamides, Aldehydes, and Ammonium Acetate. ChemistrySelect 2021. [DOI: 10.1002/slct.202100534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Seung‐Hwan Shim
- School of Life Sciences and Biotechnology Korea University Seoul 02841 Republic of Korea
| | - Hyejun Park
- School of Life Sciences and Biotechnology Korea University Seoul 02841 Republic of Korea
| | | | - In Su Kim
- School of Pharmacy Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Jae Kyun Lee
- Center for Neuromedicine Korea Institute of science and Technology Seoul 02792 Republic of Korea
| | - Kiho Lee
- College of Pharmacy Korea University Seoul 2511 Republic of Korea E-mail: sejong-ro
| | - Hitesh B. Jalani
- Smart BioPharm 310-Pilotplant Incheon Techno-Park, 12-Gaetbeol-ro, Yeonsu-gu Incheon 21999 South Korea
| | - Yongseok Choi
- School of Life Sciences and Biotechnology Korea University Seoul 02841 Republic of Korea
| |
Collapse
|
15
|
Bártolo I, Santos BS, Fontinha D, Machado M, Francisco D, Sepodes B, Rocha J, Mota-Filipe H, Pinto R, Figueira ME, Barroso H, Nascimento T, Alves de Matos AP, Alves AJS, Alves NG, Simões CJV, Prudêncio M, Pinho e Melo TMVD, Taveira N. Spiro-β-lactam BSS-730A Displays Potent Activity against HIV and Plasmodium. ACS Infect Dis 2021; 7:421-434. [PMID: 33395253 DOI: 10.1021/acsinfecdis.0c00768] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The high burden of malaria and HIV/AIDS prevents economic and social progress in developing countries. A continuing need exists for development of novel drugs and treatment regimens for both diseases in order to address the tolerability and long-term safety concerns associated with current treatment options and the emergence of drug resistance. We describe new spiro-β-lactam derivatives with potent (nM) activity against HIV and Plasmodium and no activity against bacteria and yeast. The best performing molecule of the series, BSS-730A, inhibited both HIV-1 and HIV-2 replication with an IC50 of 13 ± 9.59 nM and P. berghei hepatic infection with an IC50 of 0.55 ± 0.14 μM with a clear impact on parasite development. BSS-730A was also active against the erythrocytic stages of P. falciparum, with an estimated IC50 of 0.43 ± 0.04 μM. Time-of-addition studies showed that BSS-730A potentially affects all stages of the HIV replicative cycle, suggesting a complex mechanism of action. BSS-730A was active against multidrug-resistant HIV isolates, with a median 2.4-fold higher IC50 relative to control isolates. BSS-730A was equally active against R5 and X4 HIV isolates and displayed strong synergism with the entry inhibitor AMD3100. BSS-730A is a promising candidate for development as a potential therapeutic and/or prophylactic agent against HIV and Plasmodium.
Collapse
Affiliation(s)
- Inês Bártolo
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Bruna S. Santos
- Coimbra Chemistry Centre (CQC), Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1648-028 Lisboa, Portugal
| | - Marta Machado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1648-028 Lisboa, Portugal
| | - Denise Francisco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1648-028 Lisboa, Portugal
| | - Bruno Sepodes
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - João Rocha
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Hélder Mota-Filipe
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Rui Pinto
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria E. Figueira
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Helena Barroso
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, Quinta da Granja, 2829-511 Monte da Caparica, Portugal
| | - Teresa Nascimento
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, Quinta da Granja, 2829-511 Monte da Caparica, Portugal
| | - António P. Alves de Matos
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, Quinta da Granja, 2829-511 Monte da Caparica, Portugal
| | - Américo J. S. Alves
- Coimbra Chemistry Centre (CQC), Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Nuno G. Alves
- Coimbra Chemistry Centre (CQC), Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Carlos J. V. Simões
- Coimbra Chemistry Centre (CQC), Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1648-028 Lisboa, Portugal
| | | | - Nuno Taveira
- Instituto de investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, Quinta da Granja, 2829-511 Monte da Caparica, Portugal
| |
Collapse
|
16
|
Milheiro SA, Gonçalves J, Lopes RMRM, Madureira M, Lobo L, Lopes A, Nogueira F, Fontinha D, Prudêncio M, M Piedade MF, Pinto SN, Florindo PR, Moreira R. Half-Sandwich Cyclopentadienylruthenium(II) Complexes: A New Antimalarial Chemotype. Inorg Chem 2020; 59:12722-12732. [PMID: 32838513 DOI: 10.1021/acs.inorgchem.0c01795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A small library of "half-sandwich" cyclopentadienylruthenium(II) compounds of the general formula [(η5-C5R5)Ru(PPh3)(N-N)][PF6], a scaffold hitherto absent from the toolbox of antiplasmodials, was screened for activity against the blood stage of CQ-sensitive 3D7-GFP, CQ-resistant Dd2, and artemisinin-resistant IPC5202 Plasmodium falciparum strains and the liver stage of Plasmodium berghei. The best-performing compounds displayed dual-stage activity, with single-digit nanomolar IC50 values against blood-stage malaria parasites, nanomolar activity against liver-stage parasites, and residual cytotoxicity against HepG2 and Huh7 mammalian cells. The parasitic absorption/distribution of 7-nitrobenzoxadiazole-appended fluorescent compounds Ru4 and Ru5 was investigated by confocal fluorescence microscopy, revealing parasite-selective absorption in infected erythrocytes and nuclear accumulation of both compounds. The lead compound Ru2 impaired asexual parasite differentiation, exhibiting fast parasiticidal activity against both ring and trophozoite stages of a synchronized culture of the P. falciparum 3D7 strain. These results point to cyclopentadienylruthenium(II) complexes as a highly promising chemotype for the development of dual-stage antiplasmodials.
Collapse
Affiliation(s)
- Sofia A Milheiro
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Joana Gonçalves
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ricardo M R M Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Margarida Madureira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Lis Lobo
- Department of Medical Parasitology, Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Andreia Lopes
- Department of Medical Parasitology, Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Fátima Nogueira
- Department of Medical Parasitology, Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - M Fátima M Piedade
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Sandra N Pinto
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Pedro R Florindo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
17
|
Repurposing Drugs to Fight Hepatic Malaria Parasites. Molecules 2020; 25:molecules25153409. [PMID: 32731386 PMCID: PMC7435416 DOI: 10.3390/molecules25153409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Malaria remains one of the most prevalent infectious diseases worldwide, primarily affecting some of the most vulnerable populations around the globe. Despite achievements in the treatment of this devastating disease, there is still an urgent need for the discovery of new drugs that tackle infection by Plasmodium parasites. However, de novo drug development is a costly and time-consuming process. An alternative strategy is to evaluate the anti-plasmodial activity of compounds that are already approved for other purposes, an approach known as drug repurposing. Here, we will review efforts to assess the anti-plasmodial activity of existing drugs, with an emphasis on the obligatory and clinically silent liver stage of infection. We will also review the current knowledge on the classes of compounds that might be therapeutically relevant against Plasmodium in the context of other communicable diseases that are prevalent in regions where malaria is endemic. Repositioning existing compounds may constitute a faster solution to the current gap of prophylactic and therapeutic drugs that act on Plasmodium parasites, overall contributing to the global effort of malaria eradication.
Collapse
|
18
|
Kancharla P, Dodean RA, Li Y, Pou S, Pybus B, Melendez V, Read L, Bane CE, Vesely B, Kreishman-Deitrick M, Black C, Li Q, Sciotti RJ, Olmeda R, Luong TL, Gaona H, Potter B, Sousa J, Marcsisin S, Caridha D, Xie L, Vuong C, Zeng Q, Zhang J, Zhang P, Lin H, Butler K, Roncal N, Gaynor-Ohnstad L, Leed SE, Nolan C, Ceja FG, Rasmussen SA, Tumwebaze PK, Rosenthal PJ, Mu J, Bayles BR, Cooper RA, Reynolds KA, Smilkstein MJ, Riscoe MK, Kelly JX. Lead Optimization of Second-Generation Acridones as Broad-Spectrum Antimalarials. J Med Chem 2020; 63:6179-6202. [PMID: 32390431 PMCID: PMC7354843 DOI: 10.1021/acs.jmedchem.0c00539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The global impact of malaria remains staggering despite extensive efforts to eradicate the disease. With increasing drug resistance and the absence of a clinically available vaccine, there is an urgent need for novel, affordable, and safe drugs for prevention and treatment of malaria. Previously, we described a novel antimalarial acridone chemotype that is potent against both blood-stage and liver-stage malaria parasites. Here, we describe an optimization process that has produced a second-generation acridone series with significant improvements in efficacy, metabolic stability, pharmacokinetics, and safety profiles. These findings highlight the therapeutic potential of dual-stage targeting acridones as novel drug candidates for further preclinical development.
Collapse
Affiliation(s)
- Papireddy Kancharla
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Rozalia A. Dodean
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Yuexin Li
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Sovitj Pou
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Brandon Pybus
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Victor Melendez
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Lisa Read
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Charles E. Bane
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Brian Vesely
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Mara Kreishman-Deitrick
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Chad Black
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Qigui Li
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Richard J. Sciotti
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Raul Olmeda
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Thu-Lan Luong
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Heather Gaona
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Brittney Potter
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Jason Sousa
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Sean Marcsisin
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Diana Caridha
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Lisa Xie
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Chau Vuong
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Qiang Zeng
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Jing Zhang
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Ping Zhang
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Hsiuling Lin
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kirk Butler
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Norma Roncal
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Lacy Gaynor-Ohnstad
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Susan E. Leed
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Christina Nolan
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Frida G. Ceja
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, United States
| | - Stephanie A. Rasmussen
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, United States
| | | | - Philip J. Rosenthal
- Department of Medicine, University of California, San Francisco, CA 94143, United States
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD 20852, USA
| | - Brett R. Bayles
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, United States
- Global Public Health Program, Dominican University of California, San Rafael CA 94901
| | - Roland A. Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, United States
| | - Kevin A. Reynolds
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Martin J. Smilkstein
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Michael K. Riscoe
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Jane X. Kelly
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| |
Collapse
|
19
|
Tiwari MK, Chaudhary S. Artemisinin-derived antimalarial endoperoxides from bench-side to bed-side: Chronological advancements and future challenges. Med Res Rev 2020; 40:1220-1275. [PMID: 31930540 DOI: 10.1002/med.21657] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/21/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022]
Abstract
According to WHO World Malaria Report (2018), nearly 219 million new cases of malaria occurred and a total no. of 435 000 people died in 2017 due to this infectious disease. This is due to the rapid spread of parasite-resistant strains. Artemisinin (ART), a sesquiterpene lactone endoperoxide isolated from traditional Chinese herb Artemisia annua, has been recognized as a novel class of antimalarial drugs. The 2015 "Nobel Prize in Physiology or Medicine" was given to Prof Dr Tu Youyou for the discovery of ART. Hence, ART is termed as "Nobel medicine." The present review article accommodates insights from the chronological advancements and direct statistics witnessed during the past 48 years (1971-2019) in the medicinal chemistry of ART-derived antimalarial endoperoxides, and their clinical utility in malaria chemotherapy and drug discovery.
Collapse
Affiliation(s)
- Mohit K Tiwari
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology Jaipur, Jaipur, India
| | - Sandeep Chaudhary
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology Jaipur, Jaipur, India
| |
Collapse
|
20
|
Narula AK, Azad CS, Nainwal LM. New dimensions in the field of antimalarial research against malaria resurgence. Eur J Med Chem 2019; 181:111353. [DOI: 10.1016/j.ejmech.2019.05.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
21
|
Boniface PK, Ferreira EI. Flavonoids as efficient scaffolds: Recent trends for malaria, leishmaniasis, Chagas disease, and dengue. Phytother Res 2019; 33:2473-2517. [PMID: 31441148 DOI: 10.1002/ptr.6383] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/04/2019] [Accepted: 04/13/2019] [Indexed: 12/21/2022]
Abstract
Endemic in 149 tropical and subtropical countries, neglected tropical diseases (NTDs) affect more than 1 billion people annually with over 500,000 deaths. Among the NTDs, some of the most severe consist of leishmaniasis, Chagas disease, and dengue. The impact of the combined NTDs closely rivals that of malaria. According to the World Health Organization, 216 million cases of malaria were reported in 2016 with 445,000 deaths. Current treatment options are associated with various limitations including widespread drug resistance, severe adverse effects, lengthy treatment duration, unfavorable toxicity profiles, and complicated drug administration procedures. Flavonoids are a class of compounds that has been the subject of considerable scientific interest. New developments of flavonoids have made promising advances for the potential treatment of malaria, leishmaniasis, Chagas disease, and dengue, with less toxicity, high efficacy, and improved bioavailability. This review summarizes the current standings of the use of flavonoids to treat malaria and neglected diseases such as leishmaniasis, Chagas disease, and dengue. Natural and synthetic flavonoids are leading compounds that can be used for developing antiprotozoal and antiviral agents. However, detailed studies on toxicity, pharmacokinetics, and mechanisms of action of these compounds are required to confirm the in vitro pharmacological claims of flavonoids for pharmaceutical applications. HIGHLIGHTS: In the current review, we have tried to compile recent discoveries on natural and synthetic flavonoids as well as their implication in the treatment of malaria, leishmaniasis, Chagas disease, and dengue. A total of 373 (220 natural and 153 synthetic) flavonoids have been evaluated for antimalarial, antileishmanial, antichagasic, and antidengue activities. Most of these flavonoids showed promising results against the above diseases. Reports on molecular modeling of flavonoid compounds to the disease target indicated encouraging results. Flavonoids can be prospected as potential leads for drug development; however, more rigorously designed studies on toxicity and pharmacokinetics, as well as the quantitative structure-activity relationship studies of these compounds, need to be addressed.
Collapse
Affiliation(s)
- Pone Kamdem Boniface
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elizabeth Igne Ferreira
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
22
|
Tiwari MK, Yadav DK, Chaudhary S. Recent Developments in Natural Product Inspired Synthetic 1,2,4- Trioxolanes (Ozonides): An Unusual Entry into Antimalarial Chemotherapy. Curr Top Med Chem 2019; 19:831-846. [DOI: 10.2174/1568026619666190412104042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 11/22/2022]
Abstract
According to WHO “World health statistics 2018”, malaria alongside acute respiratory infections
and diarrhoea, is one of the major infectious disease causing children’s death in between the
age of 1-5 years. Similarly, according to another report (2016) malaria accounts for approximately
3.14% of the total disease burden worldwide. Although malaria has been widely eradicated in many
parts of the world, the global number of cases continues to rise due to the rapid spread of malaria parasites
that are resistant to antimalarial drugs. Artemisinin (8), a major breakthrough in the antimalarial
chemotherapy was isolated from the plant Artemisia annua in 1972. Its semi-synthetic derivatives such
as artemether (9), arteether (10), and artesunic acid (11) are quite effective against multi-drug resistant
malaria strains and are currently the drug of choice for the treatment of malaria. Inspite of exhibiting
excellent antimalarial activity by artemisinin (8) and its derivatives, parallel programmes for the discovery
of novel natural and synthetic peroxides were also the area of investigation of medicinal chemists
all over the world. In these continuous efforts of extensive research, natural ozonide (1,2,4-
trioxolane) was isolated from Adiantum monochlamys (Pteridaceae) and Oleandra wallichii (Davalliaceae)
in 1976. These naturally occurring stable ozonides inspired chemists to investigate this novel
class for antimalarial chemotherapy. The first identification of unusually stable synthetic antimalarial
1,2,4-trioxolanes was reported in 1992. Thus, an unusual entry of ozonides in the field of antimalarial
chemotherapy had occurred in the early nineties. This review highlights the recent advancements and
historical developments observed during the past 42 years (1976-2018) focusing mainly on important
ventures of the antimalarial 1,2,4-trioxolanes (ozonides).
Collapse
Affiliation(s)
- Mohit K. Tiwari
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology Jaipur, Jawaharlal Nehru Marg, Jaipur-302017, India
| | - Dharmendra K. Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro 191, Yeonsu-gu, Incheon city, 406-799, Korea
| | - Sandeep Chaudhary
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology Jaipur, Jawaharlal Nehru Marg, Jaipur-302017, India
| |
Collapse
|
23
|
Raphemot R, Toro-Moreno M, Lu KY, Posfai D, Derbyshire ER. Discovery of Druggable Host Factors Critical to Plasmodium Liver-Stage Infection. Cell Chem Biol 2019; 26:1253-1262.e5. [PMID: 31257182 DOI: 10.1016/j.chembiol.2019.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/06/2019] [Accepted: 05/22/2019] [Indexed: 11/26/2022]
Abstract
Plasmodium parasites undergo an obligatory and asymptomatic developmental stage within the liver before infecting red blood cells to cause malaria. The hijacked host pathways critical to parasite infection during this hepatic phase remain poorly understood. Here, we implemented a forward genetic screen to identify over 100 host factors within the human druggable genome that are critical to P. berghei infection in hepatoma cells. Notably, we found knockdown of genes involved in protein trafficking pathways to be detrimental to parasite infection. The disruption of protein trafficking modulators, including COPB2 and GGA1, decreases P. berghei parasite size, and an immunofluorescence study suggests that these proteins are recruited to the Plasmodium parasitophorous vacuole in infected hepatocytes. These findings reveal that various host intracellular protein trafficking pathways are subverted by Plasmodium parasites during the liver stage and provide new insights into their manipulation for growth and development.
Collapse
Affiliation(s)
- Rene Raphemot
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
| | - Maria Toro-Moreno
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
| | - Kuan-Yi Lu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Dora Posfai
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Emily Rose Derbyshire
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA.
| |
Collapse
|
24
|
Dodean RA, Kancharla P, Li Y, Melendez V, Read L, Bane CE, Vesely B, Kreishman-Deitrick M, Black C, Li Q, Sciotti RJ, Olmeda R, Luong TL, Gaona H, Potter B, Sousa J, Marcsisin S, Caridha D, Xie L, Vuong C, Zeng Q, Zhang J, Zhang P, Lin H, Butler K, Roncal N, Gaynor-Ohnstad L, Leed SE, Nolan C, Huezo SJ, Rasmussen SA, Stephens MT, Tan JC, Cooper RA, Smilkstein MJ, Pou S, Winter RW, Riscoe MK, Kelly JX. Discovery and Structural Optimization of Acridones as Broad-Spectrum Antimalarials. J Med Chem 2019; 62:3475-3502. [PMID: 30852885 DOI: 10.1021/acs.jmedchem.8b01961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Malaria remains one of the deadliest diseases in the world today. Novel chemoprophylactic and chemotherapeutic antimalarials are needed to support the renewed eradication agenda. We have discovered a novel antimalarial acridone chemotype with dual-stage activity against both liver-stage and blood-stage malaria. Several lead compounds generated from structural optimization of a large library of novel acridones exhibit efficacy in the following systems: (1) picomolar inhibition of in vitro Plasmodium falciparum blood-stage growth against multidrug-resistant parasites; (2) curative efficacy after oral administration in an erythrocytic Plasmodium yoelii murine malaria model; (3) prevention of in vitro Plasmodium berghei sporozoite-induced development in human hepatocytes; and (4) protection of in vivo P. berghei sporozoite-induced infection in mice. This study offers the first account of liver-stage antimalarial activity in an acridone chemotype. Details of the design, chemistry, structure-activity relationships, safety, metabolic/pharmacokinetic studies, and mechanistic investigation are presented herein.
Collapse
Affiliation(s)
- Rozalia A Dodean
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Papireddy Kancharla
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States
| | - Yuexin Li
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Victor Melendez
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Lisa Read
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Charles E Bane
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Brian Vesely
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Mara Kreishman-Deitrick
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Chad Black
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Qigui Li
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Richard J Sciotti
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Raul Olmeda
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Thu-Lan Luong
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Heather Gaona
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Brittney Potter
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Jason Sousa
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Sean Marcsisin
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Diana Caridha
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Lisa Xie
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Chau Vuong
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Qiang Zeng
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Jing Zhang
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Ping Zhang
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Hsiuling Lin
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Kirk Butler
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Norma Roncal
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Lacy Gaynor-Ohnstad
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Susan E Leed
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Christina Nolan
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Stephanie J Huezo
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | - Stephanie A Rasmussen
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | | | | | - Roland A Cooper
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | - Martin J Smilkstein
- Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Sovitj Pou
- Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Rolf W Winter
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Michael K Riscoe
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Jane X Kelly
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| |
Collapse
|
25
|
Singh S, Rajendran V, He J, Singh AK, Achieng AO, Vandana, Pant A, Nasamu AS, Pandit M, Singh J, Quadiri A, Gupta N, Poonam, Ghosh PC, Singh BK, Narayanan L, Kempaiah P, Chandra R, Dunn BM, Pandey KC, Goldberg DE, Singh AP, Rathi B. Fast-Acting Small Molecules Targeting Malarial Aspartyl Proteases, Plasmepsins, Inhibit Malaria Infection at Multiple Life Stages. ACS Infect Dis 2019; 5:184-198. [PMID: 30554511 DOI: 10.1021/acsinfecdis.8b00197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The eradication of malaria remains challenging due to the complex life cycle of Plasmodium and the rapid emergence of drug-resistant forms of Plasmodium falciparum and Plasmodium vivax. New, effective, and inexpensive antimalarials against multiple life stages of the parasite are urgently needed to combat the spread of malaria. Here, we synthesized a set of novel hydroxyethylamines and investigated their activities in vitro and in vivo. All of the compounds tested had an inhibitory effect on the blood stage of P. falciparum at submicromolar concentrations, with the best showing 50% inhibitory concentrations (IC50) of around 500 nM against drug-resistant P. falciparum parasites. These compounds showed inhibitory actions against plasmepsins, a family of malarial aspartyl proteases, and exhibited a marked killing effect on blood stage Plasmodium. In chloroquine-resistant Plasmodium berghei and P. berghei ANKA infected mouse models, treating mice with both compounds led to a significant decrease in blood parasite load. Importantly, two of the compounds displayed an inhibitory effect on the gametocyte stages (III-V) of P. falciparum in culture and the liver-stage infection of P. berghei both in in vitro and in vivo. Altogether, our findings suggest that fast-acting hydroxyethylamine-phthalimide analogs targeting multiple life stages of the parasite could be a valuable chemical lead for the development of novel antimalarial drugs.
Collapse
Affiliation(s)
- Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Vinoth Rajendran
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | - Jiang He
- Institute for Medical Engineering and Science, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Amit K. Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Angela O. Achieng
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
| | - Vandana
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
| | - Akansha Pant
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
| | - Armiyaw S. Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Mansi Pandit
- Bioinformatics Infrastructure Facility, Sri Venkateswara College, University of Delhi South Campus, New Delhi 110021, India
| | - Jyoti Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Afshana Quadiri
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Nikesh Gupta
- Special Centre for Nanosciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi North Campus, Delhi 110007, India
| | - Prahlad C. Ghosh
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | | | - Latha Narayanan
- Bioinformatics Infrastructure Facility, Sri Venkateswara College, University of Delhi South Campus, New Delhi 110021, India
| | - Prakasha Kempaiah
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
- Department of Medicine, Loyola University Stritch School of Medicine, 2160 South First Avenue, Chicago, Illinois 60153, United States
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Ben M. Dunn
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, Florida 32610, United States
| | - Kailash C. Pandey
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
- Department of Biochemistry, National Institute for Research in Environmental Health, ICMR, Bhopal 462001, India
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Agam P. Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
26
|
Kumari A, Karnatak M, Singh D, Shankar R, Jat JL, Sharma S, Yadav D, Shrivastava R, Verma VP. Current scenario of artemisinin and its analogues for antimalarial activity. Eur J Med Chem 2018; 163:804-829. [PMID: 30579122 DOI: 10.1016/j.ejmech.2018.12.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 11/17/2022]
Abstract
Human malaria, one of the most striking, reemerging infectious diseases, is caused by several types of Plasmodium parasites. Whilst advances have been made in lowering the numbers of cases and deaths, it is clear that a strategy based solely on disease control year on year, without reducing transmission and ultimately eradicating the parasite, is unsustainable. Natural products have served as a template for the design and development of antimalarial drugs currently in the clinic or in the development phase. Artemisinin combine potent, rapid antimalarial activity with a wide therapeutic index and an absence of clinically important resistance. The alkylating ability of artemisinin and its semi-synthetic analogues toward heme related to their antimalarial efficacy are underlined. Although impressive results have already been achieved in malaria research, more systematization and concentration of efforts are required if real breakthroughs are to be made. This review will concisely cover the clinical, preclinical antimalarial and current updates in artemisinin based antimalarial drugs. Diverse classes of semi-synthetic analogs of artemisinin reported in the last decade have also been extensively studied. The experience gained in this respect is discussed.
Collapse
Affiliation(s)
- Akriti Kumari
- Department of Chemistry, Banasthali University, Banasthali Newai, 304022, Rajasthan, India
| | - Manvika Karnatak
- Department of Chemistry, Banasthali University, Banasthali Newai, 304022, Rajasthan, India
| | - Davinder Singh
- Bio-Organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, Jammu and Kashmir, India
| | - Ravi Shankar
- Bio-Organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, Jammu and Kashmir, India
| | - Jawahar L Jat
- Department of Applied Chemistry, BabaSaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar Raebareli Road, Lucknow, 226025, India
| | - Siddharth Sharma
- Department of Chemistry, Mohanlal Sukhadia University, Udaipur, 313001, India
| | - Dinesh Yadav
- Department of Chemistry, Mohanlal Sukhadia University, Udaipur, 313001, India
| | - Rahul Shrivastava
- Department of Chemistry, Manipal University Jaipur, Jaipur, 303007, India
| | - Ved Prakash Verma
- Department of Chemistry, Banasthali University, Banasthali Newai, 304022, Rajasthan, India.
| |
Collapse
|
27
|
Capela R, Magalhães J, Miranda D, Machado M, Sanches-Vaz M, Albuquerque IS, Sharma M, Gut J, Rosenthal PJ, Frade R, Perry MJ, Moreira R, Prudêncio M, Lopes F. Endoperoxide-8-aminoquinoline hybrids as dual-stage antimalarial agents with enhanced metabolic stability. Eur J Med Chem 2018; 149:69-78. [PMID: 29499488 DOI: 10.1016/j.ejmech.2018.02.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/28/2018] [Accepted: 02/14/2018] [Indexed: 11/29/2022]
Abstract
Hybrid compounds may play a critical role in the context of the malaria eradication agenda, which will benefit from therapeutic tools active against the symptomatic erythrocytic stage of Plasmodium infection, and also capable of eliminating liver stage parasites. To address the need for efficient multistage antiplasmodial compounds, a small library of 1,2,4,5-tetraoxane-8- aminoquinoline hybrids, with the metabolically labile C-5 position of the 8-aminoquinoline moiety blocked with aryl groups, was synthesized and screened for antiplasmodial activity and metabolic stability. The hybrid compounds inhibited development of intra-erythrocytic forms of the multidrug-resistant Plasmodium falciparum W2 strain, with EC50 values in the nM range, and with low cytotoxicity against mammalian cells. The compounds also inhibited the development of P. berghei liver stage parasites, with the most potent compounds displaying EC50 values in the low μM range. SAR analysis revealed that unbranched linkers between the endoperoxide and 8-aminoquinoline pharmacophores are most beneficial for dual antiplasmodial activity. Importantly, hybrids were significantly more potent than a 1:1 mixture of 8-aminoquinoline-tetraoxane, highlighting the superiority of the hybrid approach over the combination therapy. Furthermore, aryl substituents at C-5 of the 8-aminoquinoline moiety improve the compounds' metabolic stability when compared with their primaquine (i.e. C-5 unsubstituted) counterparts. Overall, this study reveals that blocking the quinoline C-5 position does not result in loss of dual-stage antimalarial activity, and that tetraoxane-8- aminoquinoline hybrids are an attractive approach to achieve elimination of exo- and intraerythrocytic parasites, thus with the potential to be used in malaria eradication campaigns.
Collapse
Affiliation(s)
- Rita Capela
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Joana Magalhães
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Daniela Miranda
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Marta Machado
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Margarida Sanches-Vaz
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Inês S Albuquerque
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Moni Sharma
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Jiri Gut
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, Box 0811, CA 94143, USA
| | - Philip J Rosenthal
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, Box 0811, CA 94143, USA
| | - Raquel Frade
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria J Perry
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028, Lisboa, Portugal.
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
28
|
Poonam, Gupta Y, Gupta N, Singh S, Wu L, Chhikara BS, Rawat M, Rathi B. Multistage inhibitors of the malaria parasite: Emerging hope for chemoprotection and malaria eradication. Med Res Rev 2018; 38:1511-1535. [DOI: 10.1002/med.21486] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/09/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Poonam
- Department of Chemistry; Miranda House, University of Delhi; India
| | - Yash Gupta
- National Institute of Malaria Research (ICMR); New Delhi India
| | - Nikesh Gupta
- Special Centre for Nanosciences; Jawaharlal Nehru University; New Delhi India
| | - Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry; Hansraj College University Enclave, University of Delhi; Delhi India
| | - Lidong Wu
- Department of Chemistry; Massachusetts Institute of Technology; Cambridge MA USA
- Key Laboratory of Control of Quality and Safety for Aquatic Products; Ministry of Agriculture, Chinese Academy of Fishery Sciences; Beijing China
| | | | - Manmeet Rawat
- Department of Internal Medicine; University of New Mexico School of Medicine; Albuquerque NM USA
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry; Hansraj College University Enclave, University of Delhi; Delhi India
- Department of Chemistry; Massachusetts Institute of Technology; Cambridge MA USA
| |
Collapse
|
29
|
Nyboer B, Heiss K, Mueller AK, Ingmundson A. The Plasmodium liver-stage parasitophorous vacuole: A front-line of communication between parasite and host. Int J Med Microbiol 2017; 308:107-117. [PMID: 28964681 DOI: 10.1016/j.ijmm.2017.09.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/19/2017] [Accepted: 09/11/2017] [Indexed: 12/13/2022] Open
Abstract
The intracellular development and differentiation of the Plasmodium parasite in the host liver is a prerequisite for the actual onset of malaria disease pathology. Since liver-stage infection is clinically silent and can be completely eliminated by sterilizing immune responses, it is a promising target for urgently needed innovative antimalarial drugs and/or vaccines. Discovered more than 65 years ago, these stages remain poorly understood regarding their molecular repertoire and interaction with their host cells in comparison to the pathogenic erythrocytic stages. The differentiating and replicative intrahepatic parasite resides in a membranous compartment called the parasitophorous vacuole, separating it from the host-cell cytoplasm. Here we outline seminal work that contributed to our present understanding of the fundamental dynamic cellular processes of the intrahepatic malarial parasite with both specific host-cell factors and compartments.
Collapse
Affiliation(s)
- Britta Nyboer
- Centre for Infectious Diseases, Parasitology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Kirsten Heiss
- Centre for Infectious Diseases, Parasitology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Centre for Infection Research (DZIF), D 69120 Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Centre for Infection Research (DZIF), D 69120 Heidelberg, Germany,.
| | - Alyssa Ingmundson
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, Philippstrasse 13, 10115 Berlin, Germany.
| |
Collapse
|
30
|
Machado M, Sanches-Vaz M, Cruz JP, Mendes AM, Prudêncio M. Inhibition of Plasmodium Hepatic Infection by Antiretroviral Compounds. Front Cell Infect Microbiol 2017; 7:329. [PMID: 28770176 PMCID: PMC5515864 DOI: 10.3389/fcimb.2017.00329] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/03/2017] [Indexed: 11/13/2022] Open
Abstract
Recent WHO guidelines on control of human immunodeficiency virus (HIV) call for the widespread use of antiretroviral (AR) therapy (ART) for people living with HIV. Given the considerable overlap between infections by HIV and Plasmodium, the causative agent of malaria, it is important to understand the impact of AR compounds and ART regimens on infections by malaria parasites. We undertook a systematic approach to identify AR drugs and ART drug combinations with inhibitory activity against the obligatory hepatic stage of Plasmodium infection. Our in vitro screen of a wide array of AR drugs identified the non-nucleoside reverse transcriptase inhibitors efavirenz and etravirine (ETV), and the protease inhibitor nelfinavir, as compounds that significantly impair the development of the rodent malaria parasite P. berghei in an hepatoma cell line. Furthermore, we show that WHO-recommended ART drug combinations currently employed in the field strongly inhibit Plasmodium liver infection in mice, an effect that may be significantly enhanced by the inclusion of ETV in the treatment. Our observations are the first report of ETV as an anti-Plasmodial drug, paving the way for further evaluation and potential use of ETV-containing ARTs in regions of geographical overlap between HIV and Plasmodium infections.
Collapse
Affiliation(s)
- Marta Machado
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de LisboaLisboa, Portugal
| | - Margarida Sanches-Vaz
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de LisboaLisboa, Portugal
| | - João P Cruz
- iMed.UL-Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia da Universidade de LisboaLisboa, Portugal
| | - António M Mendes
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de LisboaLisboa, Portugal
| | - Miguel Prudêncio
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de LisboaLisboa, Portugal
| |
Collapse
|
31
|
Abstract
Prior to infecting erythrocytes and causing malaria symptoms, Plasmodium parasites undergo an obligatory phase of invasion and extensive replication inside their mammalian host's liver cells that depends on the parasite's ability to obtain the nutrients it requires for its intra-hepatic growth and multiplication. Here, we show that L-arginine (Arg) uptake through the host cell's SLC7A2-encoded transporters is essential for the parasite's development and maturation in the liver. Our data suggest that the Arg that is taken up is primarily metabolized by the arginase pathway to produce the polyamines required for Plasmodium growth. Although the parasite may hijack the host's biosynthesis pathway, it relies mainly upon its own arginase-AdoMetDC/ODC pathway to acquire the polyamines it needs to develop. These results identify for the first time a pivotal role for Arg-dependent polyamine production during Plasmodium's hepatic development and pave the way to the exploitation of strategies to impact liver infection by the malaria parasite through the modulation of Arg uptake and polyamine synthesis.
Collapse
|
32
|
Azad CS, Saxena M, Siddiqui AJ, Bhardwaj J, Puri SK, Dutta GP, Anand N, Saxena AK. Synthesis of primaquine glyco-conjugates as potential tissue schizontocidal antimalarial agents. Chem Biol Drug Des 2017; 90:254-261. [DOI: 10.1111/cbdd.12944] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Chandra S. Azad
- Division of Medicinal and Process Chemistry; CSIR-Central Drug Research Institute; Lucknow UP India
| | - Mridula Saxena
- Department of Chemistry; Amity University (Lucknow Campus); Lucknow UP India
| | - Arif J. Siddiqui
- Division of Parasitology; CSIR-Central Drug Research Institute; Lucknow UP India
| | - Jyoti Bhardwaj
- Division of Parasitology; CSIR-Central Drug Research Institute; Lucknow UP India
| | - Sunil K. Puri
- Division of Parasitology; CSIR-Central Drug Research Institute; Lucknow UP India
| | - Guru P. Dutta
- Division of Medicinal and Process Chemistry; CSIR-Central Drug Research Institute; Lucknow UP India
| | - Nitya Anand
- Division of Medicinal and Process Chemistry; CSIR-Central Drug Research Institute; Lucknow UP India
| | - Anil K. Saxena
- Division of Medicinal and Process Chemistry; CSIR-Central Drug Research Institute; Lucknow UP India
| |
Collapse
|
33
|
Plasmodium berghei EXP-1 interacts with host Apolipoprotein H during Plasmodium liver-stage development. Proc Natl Acad Sci U S A 2017; 114:E1138-E1147. [PMID: 28137845 DOI: 10.1073/pnas.1606419114] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The first, obligatory replication phase of malaria parasite infections is characterized by rapid expansion and differentiation of single parasites in liver cells, resulting in the formation and release of thousands of invasive merozoites into the bloodstream. Hepatic Plasmodium development occurs inside a specialized membranous compartment termed the parasitophorous vacuole (PV). Here, we show that, during the parasite's hepatic replication, the C-terminal region of the parasitic PV membrane protein exported protein 1 (EXP-1) binds to host Apolipoprotein H (ApoH) and that this molecular interaction plays a pivotal role for successful Plasmodium liver-stage development. Expression of a truncated EXP-1 protein, missing the specific ApoH interaction site, or down-regulation of ApoH expression in either hepatic cells or mouse livers by RNA interference resulted in impaired intrahepatic development. Furthermore, infection of mice with sporozoites expressing a truncated version of EXP-1 resulted in both a significant reduction of liver burden and delayed blood-stage patency, leading to a disease outcome different from that generally induced by infection with wild-type parasites. This study identifies a host-parasite protein interaction during the hepatic stage of infection by Plasmodium parasites. The identification of such vital interactions may hold potential toward the development of novel malaria prevention strategies.
Collapse
|
34
|
Inhibition of Plasmodium Liver Infection by Ivermectin. Antimicrob Agents Chemother 2017; 61:AAC.02005-16. [PMID: 27895022 PMCID: PMC5278742 DOI: 10.1128/aac.02005-16] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/18/2016] [Indexed: 12/29/2022] Open
Abstract
Avermectins are powerful endectocides with an established potential to reduce the incidence of vector-borne diseases. Here, we show that several avermectins inhibit the hepatic stage of Plasmodium infection in vitro. Notably, ivermectin potently inhibits liver infection in vivo by impairing parasite development inside hepatocytes. This impairment has a clear impact on the ensuing blood stage parasitemia, reducing disease severity and enhancing host survival. Ivermectin has been proposed as a tool to control malaria transmission because of its effects on the mosquito vector. Our study extends the effect of ivermectin to the early stages of mammalian host infection and supports the inclusion of this multipurpose drug in malaria control strategies.
Collapse
|
35
|
Chloroquine-containing organoruthenium complexes are fast-acting multistage antimalarial agents. Parasitology 2016; 143:1543-56. [PMID: 27439976 DOI: 10.1017/s0031182016001153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We report the pharmacological activity of organoruthenium complexes containing chloroquine (CQ) as a chelating ligand. The complexes displayed intraerythrocytic activity against CQ-sensitive 3D7 and CQ-resistant W2 strains of Plasmodium falciparum, with potency and selectivity indexes similar to those of CQ. Complexes displayed activity against all intraerythrocytic stages, but moderate activity against Plasmodium berghei liver stages. However, unlike CQ, organoruthenium complexes impaired gametocyte viability and exhibited fast parasiticidal activity against trophozoites for P. falciparum. This functional property results from the ability of complexes to quickly induce oxidative stress. The parasitaemia of P. berghei-infected mice was reduced by treatment with the complex. Our findings demonstrated that using chloroquine for the synthesis of organoruthenium complexes retains potency and selectivity while leading to an increase in the spectrum of action and parasite killing rate relative to CQ.
Collapse
|
36
|
Raphemot R, Posfai D, Derbyshire ER. Current therapies and future possibilities for drug development against liver-stage malaria. J Clin Invest 2016; 126:2013-20. [PMID: 27249674 DOI: 10.1172/jci82981] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Malaria remains a global public health threat, with half of the world's population at risk. Despite numerous efforts in the past decade to develop new antimalarial drugs to surmount increasing resistance to common therapies, challenges remain in the expansion of the current antimalarial arsenal for the elimination of this disease. The requirement of prophylactic and radical cure activities for the next generation of antimalarial drugs demands that new research models be developed to support the investigation of the elusive liver stage of the malaria parasite. In this Review, we revisit current antimalarial therapies and discuss recent advances for in vitro and in vivo malaria research models of the liver stage and their importance in probing parasite biology and the discovery of novel drug candidates.
Collapse
|
37
|
Hentzschel F, Herrmann AK, Mueller AK, Grimm D. Plasmodium meets AAV-the (un)likely marriage of parasitology and virology, and how to make the match. FEBS Lett 2016; 590:2027-45. [PMID: 27117587 DOI: 10.1002/1873-3468.12187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/24/2016] [Accepted: 04/21/2016] [Indexed: 01/21/2023]
Abstract
The increasing use of screening technologies in malaria research has substantially expanded our knowledge on cellular factors hijacked by the Plasmodium parasite in the infected host, including those that participate in the clinically silent liver stage. This rapid gain in our understanding of the hepatic interaction partners now requires a means to validate and further disentangle parasite-host networks in physiologically relevant liver model systems. Here, we outline seminal work that contributed to our present knowledge on the intrahepatic Plasmodium host factors, followed by a discussion of surrogate models of mammalian livers or hepatocytes. We finally describe how Adeno-associated viruses could be engineered and used as hepatotropic tools to dissect Plasmodium-host interactions, and to deliberately control these networks for antimalaria vaccination or therapy.
Collapse
Affiliation(s)
- Franziska Hentzschel
- Department of Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Anne-Kathrin Herrmann
- Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Department of Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Germany
| | - Dirk Grimm
- Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Cluster of Excellence CellNetworks, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| |
Collapse
|
38
|
Ribeiro CJ, Espadinha M, Machado M, Gut J, Gonçalves LM, Rosenthal PJ, Prudêncio M, Moreira R, Santos MM. Novel squaramides with in vitro liver stage antiplasmodial activity. Bioorg Med Chem 2016; 24:1786-92. [DOI: 10.1016/j.bmc.2016.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/02/2016] [Indexed: 12/17/2022]
|
39
|
Terzić N, Konstantinović J, Tot M, Burojević J, Djurković-Djaković O, Srbljanović J, Štajner T, Verbić T, Zlatović M, Machado M, Albuquerque IS, Prudêncio M, Sciotti RJ, Pecic S, D'Alessandro S, Taramelli D, Šolaja BA. Reinvestigating Old Pharmacophores: Are 4-Aminoquinolines and Tetraoxanes Potential Two-Stage Antimalarials? J Med Chem 2015; 59:264-81. [PMID: 26640981 DOI: 10.1021/acs.jmedchem.5b01374] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The syntheses and antiplasmodial activities of various substituted aminoquinolines coupled to an adamantane carrier are described. The compounds exhibited pronounced in vitro and in vivo activity against Plasmodium berghei in the Thompson test. Tethering a fluorine atom to the aminoquinoline C(3) position afforded fluoroaminoquinolines that act as intrahepatocytic parasite inhibitors, with compound 25 having an IC50 = 0.31 μM and reducing the liver load in mice by up to 92% at 80 mg/kg dose. Screening our peroxides as inhibitors of liver stage infection revealed that the tetraoxane pharmacophore itself is also an excellent liver stage P. berghei inhibitor (78: IC50 = 0.33 μM). Up to 91% reduction of the parasite liver load in mice was achieved at 100 mg/kg. Examination of tetraoxane 78 against the transgenic 3D7 strain expressing luciferase under a gametocyte-specific promoter revealed its activity against stage IV-V Plasmodium falciparum gametocytes (IC50 = 1.16 ± 0.37 μM). To the best of our knowledge, compounds 25 and 78 are the first examples of either an 4-aminoquinoline or a tetraoxane liver stage inhibitors.
Collapse
Affiliation(s)
- Natasa Terzić
- Institute of Chemistry, Technology, and Metallurgy , 11000 Belgrade, Serbia
| | - Jelena Konstantinović
- Faculty of Chemistry, University of Belgrade , Studentski trg 16, P.O. Box 51, 11158, Belgrade, Serbia
| | - Mikloš Tot
- Faculty of Chemistry, University of Belgrade , Studentski trg 16, P.O. Box 51, 11158, Belgrade, Serbia
| | - Jovana Burojević
- Faculty of Chemistry, University of Belgrade , Studentski trg 16, P.O. Box 51, 11158, Belgrade, Serbia
| | | | - Jelena Srbljanović
- Institute for Medical Research, University of Belgrade , Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Tijana Štajner
- Institute for Medical Research, University of Belgrade , Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Tatjana Verbić
- Faculty of Chemistry, University of Belgrade , Studentski trg 16, P.O. Box 51, 11158, Belgrade, Serbia
| | - Mario Zlatović
- Faculty of Chemistry, University of Belgrade , Studentski trg 16, P.O. Box 51, 11158, Belgrade, Serbia
| | - Marta Machado
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , 1649-028 Lisboa, Portugal
| | - Inês S Albuquerque
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , 1649-028 Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , 1649-028 Lisboa, Portugal
| | - Richard J Sciotti
- Experimental Therapeutics Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland 20910, United States
| | - Stevan Pecic
- Division of Experimental Therapeutics, Department of Medicine, Columbia University , New York, New York 10032, United States
| | - Sarah D'Alessandro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano , 20133 Milan, Italy
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano , 20133 Milan, Italy
| | - Bogdan A Šolaja
- Faculty of Chemistry, University of Belgrade , Studentski trg 16, P.O. Box 51, 11158, Belgrade, Serbia
| |
Collapse
|
40
|
Kaur H, Machado M, de Kock C, Smith P, Chibale K, Prudêncio M, Singh K. Primaquine-pyrimidine hybrids: synthesis and dual-stage antiplasmodial activity. Eur J Med Chem 2015; 101:266-73. [PMID: 26142491 DOI: 10.1016/j.ejmech.2015.06.045] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/21/2015] [Accepted: 06/22/2015] [Indexed: 10/23/2022]
Abstract
A series of novel pyrimidine-primaquine hybrids were synthesized and their effectiveness against the blood and liver stages of malaria parasites was evaluated. The hybrids displayed enhanced liver stage in vitro activity against P. berghei liver stage infection.
Collapse
Affiliation(s)
- Hardeep Kaur
- Department of Chemistry, UGC-Centre for Advanced Studies-II, Guru Nanak Dev University, Amritsar 143005, India
| | - Marta Machado
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carmen de Kock
- Division of Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Peter Smith
- Division of Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Kelly Chibale
- Department of Chemistry, South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 701, South Africa
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Kamaljit Singh
- Department of Chemistry, UGC-Centre for Advanced Studies-II, Guru Nanak Dev University, Amritsar 143005, India.
| |
Collapse
|
41
|
Campo B, Vandal O, Wesche DL, Burrows JN. Killing the hypnozoite--drug discovery approaches to prevent relapse in Plasmodium vivax. Pathog Glob Health 2015; 109:107-22. [PMID: 25891812 PMCID: PMC4455353 DOI: 10.1179/2047773215y.0000000013] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The eradication of malaria will only be possible if effective, well-tolerated medicines kill hypnozoites in vivax and ovale malaria, and thus prevent relapses in patients. Despite progress in the 8-aminoquinoline series, with tafenoquine in Phase III showing clear benefits over primaquine, the drug discovery challenge to identify hypnozoiticidal or hypnozoite-activating compounds has been hampered by the dearth of biological tools and assays, which in turn has been limited by the immense scientific and logistical challenges associated with accessing relevant human tissue and sporozoites. This review summarises the existing drug discovery series and approaches concerning the goal to block relapse.
Collapse
Affiliation(s)
- Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Omar Vandal
- The Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - David L. Wesche
- The Bill and Melinda Gates Foundation, Seattle, WA, USA
- Great Lakes Drug Development/Certara, Princeton, NJ, USA
| | | |
Collapse
|
42
|
Cohen A, Suzanne P, Lancelot JC, Verhaeghe P, Lesnard A, Basmaciyan L, Hutter S, Laget M, Dumètre A, Paloque L, Deharo E, Crozet MD, Rathelot P, Dallemagne P, Lorthiois A, Sibley CH, Vanelle P, Valentin A, Mazier D, Rault S, Azas N. Discovery of new thienopyrimidinone derivatives displaying antimalarial properties toward both erythrocytic and hepatic stages of Plasmodium. Eur J Med Chem 2015; 95:16-28. [DOI: 10.1016/j.ejmech.2015.03.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 11/24/2022]
|
43
|
From hybrid compounds to targeted drug delivery in antimalarial therapy. Bioorg Med Chem 2015; 23:5120-30. [PMID: 25913864 DOI: 10.1016/j.bmc.2015.04.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 11/21/2022]
Abstract
The discovery of new drugs to treat malaria is a continuous effort for medicinal chemists due to the emergence and spread of resistant strains of Plasmodium falciparum to nearly all used antimalarials. The rapid adaptation of the malaria parasite remains a major limitation to disease control. Development of hybrid antimalarial agents has been actively pursued as a promising strategy to overcome the emergence of resistant parasite strains. This review presents the journey that started with simple combinations of two active moieties into one chemical entity and progressed into a delivery/targeted system based on major antimalarial classes of drugs. The rationale for providing different mechanisms of action against a single or additional targets involved in the multiple stages of the parasite's life-cycle is highlighted. Finally, a perspective for this polypharmacologic approach is presented.
Collapse
|
44
|
Rodrigues CAB, Frade RFM, Albuquerque IS, Perry MJ, Gut J, Machado M, Rosenthal PJ, Prudêncio M, Afonso CAM, Moreira R. Targeting the Erythrocytic and Liver Stages of Malaria Parasites withs-Triazine-Based Hybrids. ChemMedChem 2015; 10:883-90. [DOI: 10.1002/cmdc.201500011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/15/2015] [Indexed: 11/08/2022]
|
45
|
Agarwal D, Sharma M, Dixit SK, Dutta RK, Singh AK, Gupta RD, Awasthi SK. In vitro synergistic effect of fluoroquinolone analogues in combination with artemisinin against Plasmodium falciparum; their antiplasmodial action in rodent malaria model. Malar J 2015; 14:48. [PMID: 25652883 PMCID: PMC4349624 DOI: 10.1186/s12936-015-0561-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/14/2015] [Indexed: 11/10/2022] Open
Abstract
Background Emergence of drug-resistant parasite strains has surfaced as a major obstacle in attempts to ameliorate malaria. Current treatment regimen of malaria relies on the concept of artemisinin-based combination therapy (ACT). Methods Fluoroquinolone analogues, compounds 10, 12 and 18 were investigated for their anti-malarial interaction in combination with artemisinin in vitro, against Plasmodium falciparum 3D7 strain, employing fixed-ratio combination isobologram method. In addition, the efficacy of these compounds was evaluated intraperitoneally in BALB/c mice infected with chloroquine-resistant Plasmodium berghei ANKA strain in the Peters’ four-day suppressive test. Results Promising results were obtained in the form of synergistic or additive interactions. Compounds 10 and 12 were found to have highly synergistic interactions with artemisinin. Antiplasmodial effect was further verified by the convincing ED50 values of these compounds, which ranged between 2.31 and 3.09 (mg/kg BW). Conclusions In vivo studies substantiated the potential of the fluoroquinolone derivatives to be developed as synergistic partners for anti-malarial drug combinations.
Collapse
Affiliation(s)
- Drishti Agarwal
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India. .,Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India.
| | - Manish Sharma
- Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Sandeep K Dixit
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India.
| | - Roshan K Dutta
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India.
| | - Ashok K Singh
- Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Rinkoo D Gupta
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India.
| | - Satish K Awasthi
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
46
|
Sonawane DP, Persico M, Corbett Y, Chianese G, Di Dato A, Fattorusso C, Taglialatela-Scafati O, Taramelli D, Trombini C, Dhavale DD, Quintavalla A, Lombardo M. New antimalarial 3-methoxy-1,2-dioxanes: optimization of cellular pharmacokinetics and pharmacodynamics properties by incorporation of amino and N-heterocyclic moieties at C4. RSC Adv 2015. [DOI: 10.1039/c5ra10785g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A new series of 3-methoxy-1,2-dioxanes containing an amino moiety at C4 was synthesized, displaying nanomolar antimalarial activity without being cytotoxic.
Collapse
|
47
|
Vandekerckhove S, D'hooghe M. Quinoline-based antimalarial hybrid compounds. Bioorg Med Chem 2014; 23:5098-119. [PMID: 25593097 DOI: 10.1016/j.bmc.2014.12.018] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/03/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022]
Abstract
Quinoline-containing compounds, such as quinine and chloroquine, have a long-standing history as potent antimalarial agents. However, the increasing resistance of the Plasmodium parasite against these drugs and the lack of licensed malaria vaccines have forced chemists to develop synthetic strategies toward novel biologically active molecules. A strategy that has attracted considerable attention in current medicinal chemistry is based on the conjugation of two biologically active molecules into one hybrid compound. Since quinolines are considered to be privileged antimalarial building blocks, the synthesis of quinoline-containing antimalarial hybrids has been elaborated extensively in recent years. This review provides a literature overview of antimalarial hybrid molecules containing a quinoline core, covering publications between 2009 and 2014.
Collapse
Affiliation(s)
- Stéphanie Vandekerckhove
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium.
| |
Collapse
|
48
|
Amorim Madeira PJ, Sitoe ARF, Gonçalves D, Rodrigues T, Guedes RC, Lopes F, Moreira R, Bronze MR. Antiplasmodial drugs in the gas phase: a CID and DFT study of quinolon-4(1H)-imine derivatives. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2014; 25:1650-1661. [PMID: 25001380 DOI: 10.1007/s13361-014-0940-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/26/2014] [Accepted: 05/26/2014] [Indexed: 06/03/2023]
Abstract
The gas-phase behavior of 12 quinolon-4(1H)-imine derivatives with antiplasmodial activity was investigated using electrospray ionization tandem mass spectrometry together with collision induced dissociation and density functional theory (DFT) calculations. The most probable protonation site was predicted by calculating the proton affinity (PA) values for each possible protonation site and it was found to be the imine nitrogen for all compounds under study. Fragmentation pathways of the protonated molecules were proposed and the assignment of product ion structures was performed taking into account theoretical calculations. The nature of the quinoline substituent was found to influence the gas-phase behavior of the compounds under study. The data acquired allowed to bracket the proton affinity of the quinolin-4-imine scaffold, which can be a useful starting point to choose appropriate references for determining PA values of this scaffold.
Collapse
Affiliation(s)
- Paulo J Amorim Madeira
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003, Lisbon, Portugal,
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Dual-stage triterpenoids from an African medicinal plant targeting the malaria parasite. Bioorg Med Chem 2014; 22:3887-90. [DOI: 10.1016/j.bmc.2014.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 11/18/2022]
|
50
|
Hansen FK, Sumanadasa SDM, Stenzel K, Duffy S, Meister S, Marek L, Schmetter R, Kuna K, Hamacher A, Mordmüller B, Kassack MU, Winzeler EA, Avery VM, Andrews KT, Kurz T. Discovery of HDAC inhibitors with potent activity against multiple malaria parasite life cycle stages. Eur J Med Chem 2014; 82:204-13. [PMID: 24904967 DOI: 10.1016/j.ejmech.2014.05.050] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/12/2014] [Accepted: 05/21/2014] [Indexed: 10/25/2022]
Abstract
In this work we investigated the antiplasmodial activity of a series of HDAC inhibitors containing an alkoxyamide connecting-unit linker region. HDAC inhibitor 1a (LMK235), previously shown to be a novel and specific inhibitor of human HDAC4 and 5, was used as a starting point to rapidly construct a mini-library of HDAC inhibitors using a straightforward solid-phase supported synthesis. Several of these novel HDAC inhibitors were found to have potent in vitro activity against asexual stage Plasmodium falciparum malaria parasites. Representative compounds were shown to hyperacetylate P. falciparum histones and to inhibit deacetylase activity of recombinant PfHDAC1 and P. falciparum nuclear extracts. All compounds were also screened in vitro for activity against Plasmodium berghei exo-erythrocytic stages and selected compounds were further tested against late stage (IV and V) P. falciparum gametocytes. Of note, some compounds showed nanomolar activity against all three life cycle stages tested (asexual, exo-erythrocytic and gametocyte stages) and several compounds displayed significantly increased parasite selectivity compared to the reference HDAC inhibitor suberoylanilide hydroxamic acid (SAHA). These data suggest that it may be possible to develop HDAC inhibitors that target multiple malaria parasite life cycle stages.
Collapse
Affiliation(s)
- Finn K Hansen
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Subathdrage D M Sumanadasa
- Eskitis Institute for Drug Discovery, Don Young Road, Nathan Campus, Griffith University, QLD 4111, Australia
| | - Katharina Stenzel
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sandra Duffy
- Eskitis Institute for Drug Discovery, Don Young Road, Nathan Campus, Griffith University, QLD 4111, Australia
| | - Stephan Meister
- Department of Pediatrics, University of California, San Diego, School of Medicine, 9500 Gilman Drive 0741, La Jolla, CA 92093, USA
| | - Linda Marek
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Rebekka Schmetter
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Krystina Kuna
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Alexandra Hamacher
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Wilhelmstr. 27, 72074 Tübingen, Germany; Medical Research Laboratory, Albert Schweitzer Hospital, Lambaréné, Gabon
| | - Matthias U Kassack
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Elizabeth A Winzeler
- Department of Pediatrics, University of California, San Diego, School of Medicine, 9500 Gilman Drive 0741, La Jolla, CA 92093, USA
| | - Vicky M Avery
- Eskitis Institute for Drug Discovery, Don Young Road, Nathan Campus, Griffith University, QLD 4111, Australia
| | - Katherine T Andrews
- Eskitis Institute for Drug Discovery, Don Young Road, Nathan Campus, Griffith University, QLD 4111, Australia.
| | - Thomas Kurz
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|