1
|
Nguyen M, Paloque L, Manaranche J, Chabbert M, Hamouy A, Laurent M, Augereau JM, Claparols C, Robert A, Benoit-Vical F. Reductive Activation of Artefenomel (OZ439) by Fe(II)-Heme, Related to Its Antimalarial Activity. ACS Infect Dis 2025; 11:216-225. [PMID: 39681556 DOI: 10.1021/acsinfecdis.4c00787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The 1,2,4-trioxolane antimalarial drug, OZ439 (artefenomel), exhibits cross-resistance to artemisinins in vitro with similar survival rates of artemisinin-resistant parasites after dihydroartemisinin or OZ439 exposure, suggesting that this drug shares some mechanisms of action with artemisinins. In this way, we investigated the in vitro reductive activation of OZ439 by heme in the presence of dithionite, demonstrating the formation of covalent heme-drug adducts. However, in the presence of the biologically abundant reductant glutathione instead of dithionite, heme-drug adducts were not detected, contrary to artemisinin that efficiently alkylates heme regardless of the reductant used. Conversely, the C-centered radical of OZ439 resulting from heme-mediated activation of the drug reacts with the thiol function of glutathione, thus confirming the ability of this drug to alkylate proteins or other biological targets. So, the difference in the mechanism of action between artemisinin and OZ439 in vivo may rely on the different proportions between heme alkylation and protein alkylation.
Collapse
Affiliation(s)
- Michel Nguyen
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| | - Lucie Paloque
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| | - Jeanne Manaranche
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| | - Mickaël Chabbert
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| | - Alexandre Hamouy
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Marion Laurent
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| | - Jean-Michel Augereau
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| | - Catherine Claparols
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Chimie de Toulouse (UAR2599), Université de Toulouse, 118, Route de Narbonne, 31062 Toulouse Cedex, France
| | - Anne Robert
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
| | - Françoise Benoit-Vical
- Laboratoire de Chimie de Coordination du CNRS, LCC-CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Inserm ERL 1289 MAAP, Université de Toulouse, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex, France
| |
Collapse
|
2
|
Blank B, Gut J, Rosenthal PJ, Renslo AR. Artefenomel Regioisomer RLA-3107 Is a Promising Lead for the Discovery of Next-Generation Endoperoxide Antimalarials. ACS Med Chem Lett 2023; 14:493-498. [PMID: 37077383 PMCID: PMC10108391 DOI: 10.1021/acsmedchemlett.3c00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/30/2023] [Indexed: 04/21/2023] Open
Abstract
Clinical development of the antimalarial artefenomel was recently halted due to formulation challenges stemming from the drug's lipophilicity and low aqueous solubility. The symmetry of organic molecules is known to influence crystal packing energies and by extension solubility and dissolution rates. Here we evaluate RLA-3107, a desymmetrized, regioisomeric form of artefenomel in vitro and in vivo, finding that the regioisomer retains potent antiplasmodial activity while offering improved human microsome stability and aqueous solubility as compared to artefenomel. We also report in vivo efficacy data for artefenomel and its regioisomer across 12 different dosing regimens.
Collapse
Affiliation(s)
- Brian
R. Blank
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
| | - Jiri Gut
- Department
of Medicine, San Francisco General Hospital,
University of California, San Francisco, San Francisco, California 94143, United States
| | - Philip J. Rosenthal
- Department
of Medicine, San Francisco General Hospital,
University of California, San Francisco, San Francisco, California 94143, United States
| | - Adam R. Renslo
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
| |
Collapse
|
3
|
Müller J, Schlange C, Heller M, Uldry AC, Braga-Lagache S, Haynes RK, Hemphill A. Proteomic characterization of Toxoplasma gondii ME49 derived strains resistant to the artemisinin derivatives artemiside and artemisone implies potential mode of action independent of ROS formation. Int J Parasitol Drugs Drug Resist 2022; 21:1-12. [PMID: 36512904 PMCID: PMC9763631 DOI: 10.1016/j.ijpddr.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
The sesquiterpene lactone artemisinin and its amino-artemisinin derivatives artemiside (GC008) and artemisone (GC003) are potent antimalarials. The mode of action of artemisinins against Plasmodium sp is popularly ascribed to 'activation' of the peroxide group by heme-Fe(II) or labile Fe(II) to generate C-radicals that alkylate parasite proteins. An alternative postulate is that artemisinins elicit formation of reactive oxygen species by interfering with flavin disulfide reductases resposible for maintaining intraparasitic redox homeostasis. However, in contradistinction to the heme-activation mechanism, the amino-artemisinins are effective in vitro against non-heme-degrading apicomplexan parasites including T. gondii, with IC 50 values of 50-70 nM, and induce distinct ultrastructural alterations. However, T. gondii strains readily adapted to increased concentrations (2.5 μM) of these two compounds within few days. Thus, T. gondii strains that were resistant against artemisone and artemiside were generated by treating the T. gondii reference strain ME49 with stepwise increasing amounts of these compounds, yielding the artemisone resistant strain GC003R and the artemiside resistant strain GC008R. Differential analyses of the proteomes of these resistant strains compared to the wildtype ME49 revealed that 215 proteins were significantly downregulated in artemisone resistant tachyzoites and only 8 proteins in artemiside resistant tachyzoites as compared to their wildtype. Two proteins, namely a hypothetical protein encoded by ORF TGME49_236950, and the rhoptry neck protein RON2 encoded by ORF TGME49_300100 were downregulated in both resistant strains. Interestingly, eight proteins involved in ROS scavenging including catalase and superoxide dismutase were amongst the differentially downregulated proteins in the artemisone-resistant strain. In parallel, ROS formation was significantly enhanced in isolated tachyzoites from the artemisone resistant strain and - to a lesser extent - in tachyzoites from the artemiside resistant strain as compared to wildtype tachyzoites. These findings suggest that amino-artemisinin derivatives display a mechanism of action in T. gondii distinct from Plasmodium.
Collapse
Affiliation(s)
- Joachim Müller
- Institute of Parasitology, University of Bern, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Länggass-Strasse 122, CH-3012, Bern, Switzerland
| | - Carling Schlange
- Institute of Parasitology, University of Bern, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Länggass-Strasse 122, CH-3012, Bern, Switzerland
| | - Manfred Heller
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Sophie Braga-Lagache
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Richard K Haynes
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, 2520, South Africa
| | - Andrew Hemphill
- Institute of Parasitology, University of Bern, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Länggass-Strasse 122, CH-3012, Bern, Switzerland.
| |
Collapse
|
4
|
Ng JPL, Han Y, Yang LJ, Birkholtz LM, Coertzen D, Wong HN, Haynes RK, Coghi P, Wong VKW. Antimalarial and antitumour activities of the steroidal quinone-methide celastrol and its combinations with artemiside, artemisone and methylene blue. Front Pharmacol 2022; 13:988748. [PMID: 36120293 PMCID: PMC9479156 DOI: 10.3389/fphar.2022.988748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022] Open
Abstract
Artemisinin, isolated from the traditional Chinese medicinal plant qīng hāo 青蒿 (Artemisia annua) and its derivatives are used for treatment of malaria. With treatment failures now being recorded for the derivatives and companion drugs used in artemisinin combination therapies new drug combinations are urgently required. The amino-artemisinins artemiside and artemisone display optimal efficacies in vitro against asexual and sexual blood stages of the malaria parasite Plasmodium falciparum and are active against tumour cell lines. In continuing the evolution of combinations of the amino-artemisinins with new drugs, we examine the triterpenoid quinone methide celastrol isolated from the traditional Chinese medicinal plant léi gōng téng 雷公藤 (Tripterygium wilfordii). This compound is redox active, and has attracted considerable attention because of potent biological activities against manifold targets. We report that celastrol displays good IC50 activities ranging from 0.50–0.82 µM against drug-sensitive and resistant asexual blood stage Pf, and 1.16 and 0.28 µM respectively against immature and late stage Pf NF54 gametocytes. The combinations of celastrol with each of artemisone and methylene blue against asexual blood stage Pf are additive. Given that celastrol displays promising antitumour properties, we examined its activities alone and in combinations with amino-artemisinins against human liver HepG2 and other cell lines. IC50 values of the amino-artemisinins and celastrol against HepG2 cancer cells ranged from 0.55–0.94 µM. Whereas the amino-artemisinins displayed notable selectivities (SI > 171) with respect to normal human hepatocytes, in contrast, celastrol displayed no selectivity (SI < 1). The combinations of celastrol with artemiside or artemisone against HepG2 cells are synergistic. Given the promise of celastrol, judiciously designed formulations or structural modifications are recommended for mitigating its toxicity.
Collapse
Affiliation(s)
- Jerome P. L. Ng
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yu Han
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Li Jun Yang
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute Malaria for Sustainable Malaria Control, University of Pretoria, Hatfield, South Africa
| | - Dina Coertzen
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute Malaria for Sustainable Malaria Control, University of Pretoria, Hatfield, South Africa
| | - Ho Ning Wong
- Centre of Excellence for Pharmaceutical Sciences, School of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Richard K. Haynes
- Centre of Excellence for Pharmaceutical Sciences, School of Health Sciences, North-West University, Potchefstroom, South Africa
- *Correspondence: Richard K. Haynes, Paolo Coghi, Vincent Kam Wai Wong,
| | - Paolo Coghi
- School of Pharmacy, Macau University of Science and Technology, Macau, China
- *Correspondence: Richard K. Haynes, Paolo Coghi, Vincent Kam Wai Wong,
| | - Vincent Kam Wai Wong
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- *Correspondence: Richard K. Haynes, Paolo Coghi, Vincent Kam Wai Wong,
| |
Collapse
|
5
|
Sousa CC, Dziwornu GA, Quadros HC, Araujo-Neto JH, Chibale K, Moreira DRM. Antimalarial Pyrido[1,2- a]benzimidazoles Exert Strong Parasiticidal Effects by Achieving High Cellular Uptake and Suppressing Heme Detoxification. ACS Infect Dis 2022; 8:1700-1710. [PMID: 35848708 DOI: 10.1021/acsinfecdis.2c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pyrido[1,2-a]benzimidazoles (PBIs) are synthetic antiplasmodium agents with potent activity and are structurally differentiated from benchmark antimalarials. To study the cellular uptake of PBIs and understand the underlying phenotype of their antiplasmodium activity, their antiparasitic activities were examined in chloroquine (CQ)-susceptible and CQ-resistant Plasmodium falciparum in vitro. Moreover, drug uptake and heme detoxification suppression were examined in Plasmodium berghei-infected mice. The in vitro potency of PBIs is comparable to most 4-aminoquinolines. They have a speed of action in vitro that is superior to that of atovaquone and an ability to kill rings and trophozoites. The antiparasitic effects observed for the PBIs in cell culture and in infected mice are similar in terms of potency and efficacy and are comparable to CQ but with the added advantage of demonstrating equipotency against both CQ susceptible and resistant parasite strains. PBIs have a high rate of uptake by parasite cells and, conversely, a limited rate of uptake by host cells. The mechanism of cellular uptake of the PBIs differs from the ion-trap mechanism typically observed for 4-aminoquinolines, although they share key structural features. The high cellular uptake, attractive parasiticidal profile, and susceptibility of resistant strains to PBIs are desirable characteristics for new antimalarial agents.
Collapse
Affiliation(s)
- Caroline C Sousa
- Fundação Oswaldo Cruz (Fiocruz), Instituto Gonçalo Moniz, Salvador, 40296-710 Bahia, Brazil
| | - Godwin Akpeko Dziwornu
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Helenita C Quadros
- Fundação Oswaldo Cruz (Fiocruz), Instituto Gonçalo Moniz, Salvador, 40296-710 Bahia, Brazil
| | | | - Kelly Chibale
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Diogo R M Moreira
- Fundação Oswaldo Cruz (Fiocruz), Instituto Gonçalo Moniz, Salvador, 40296-710 Bahia, Brazil
| |
Collapse
|
6
|
Siddiqui G, Giannangelo C, De Paoli A, Schuh AK, Heimsch KC, Anderson D, Brown TG, MacRaild CA, Wu J, Wang X, Dong Y, Vennerstrom JL, Becker K, Creek DJ. Peroxide Antimalarial Drugs Target Redox Homeostasis in Plasmodium falciparum Infected Red Blood Cells. ACS Infect Dis 2022; 8:210-226. [PMID: 34985858 PMCID: PMC8762662 DOI: 10.1021/acsinfecdis.1c00550] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Plasmodium
falciparum causes the
most lethal form of malaria. Peroxide antimalarials based on artemisinin
underpin the frontline treatments for malaria, but artemisinin resistance
is rapidly spreading. Synthetic peroxide antimalarials, known as ozonides,
are in clinical development and offer a potential alternative. Here,
we used chemoproteomics to investigate the protein alkylation targets
of artemisinin and ozonide probes, including an analogue of the ozonide
clinical candidate, artefenomel. We greatly expanded the list of proteins
alkylated by peroxide antimalarials and identified significant enrichment
of redox-related proteins for both artemisinins and ozonides. Disrupted
redox homeostasis was confirmed by dynamic live imaging of the glutathione
redox potential using a genetically encoded redox-sensitive fluorescence-based
biosensor. Targeted liquid chromatography-mass spectrometry (LC-MS)-based
thiol metabolomics also confirmed changes in cellular thiol levels.
This work shows that peroxide antimalarials disproportionately alkylate
proteins involved in redox homeostasis and that disrupted redox processes
are involved in the mechanism of action of these important antimalarials.
Collapse
Affiliation(s)
- Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Amanda De Paoli
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Anna Katharina Schuh
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Kim C. Heimsch
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Dovile Anderson
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Timothy G. Brown
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Christopher A. MacRaild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Jianbo Wu
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Xiaofang Wang
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Yuxiang Dong
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Jonathan L. Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| |
Collapse
|
7
|
The Role of the Iron Protoporphyrins Heme and Hematin in the Antimalarial Activity of Endoperoxide Drugs. Pharmaceuticals (Basel) 2022; 15:ph15010060. [PMID: 35056117 PMCID: PMC8779033 DOI: 10.3390/ph15010060] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/05/2023] Open
Abstract
Plasmodium has evolved to regulate the levels and oxidative states of iron protoporphyrin IX (Fe-PPIX). Antimalarial endoperoxides such as 1,2,4-trioxane artemisinin and 1,2,4-trioxolane arterolane undergo a bioreductive activation step mediated by heme (FeII-PPIX) but not by hematin (FeIII-PPIX), leading to the generation of a radical species. This can alkylate proteins vital for parasite survival and alkylate heme into hematin–drug adducts. Heme alkylation is abundant and accompanied by interconversion from the ferrous to the ferric state, which may induce an imbalance in the iron redox homeostasis. In addition to this, hematin–artemisinin adducts antagonize the spontaneous biomineralization of hematin into hemozoin crystals, differing strikingly from artemisinins, which do not directly suppress hematin biomineralization. These hematin–drug adducts, despite being devoid of the peroxide bond required for radical-induced alkylation, are powerful antiplasmodial agents. This review addresses our current understanding of Fe-PPIX as a bioreductive activator and molecular target. A compelling pharmacological model is that by alkylating heme, endoperoxide drugs can cause an imbalance in the iron homeostasis and that the hematin–drug adducts formed have strong cytocidal effects by possibly reproducing some of the toxifying effects of free Fe-PPIX. The antiplasmodial phenotype and the mode of action of hematin–drug adducts open new possibilities for reconciliating the mechanism of endoperoxide drugs and for malaria intervention.
Collapse
|
8
|
Wittlin S, Mäser P. From Magic Bullet to Magic Bomb: Reductive Bioactivation of Antiparasitic Agents. ACS Infect Dis 2021; 7:2777-2786. [PMID: 34472830 DOI: 10.1021/acsinfecdis.1c00118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Paul Ehrlich coined the term "magic bullet" to describe how a drug kills the parasite inside its human host without harming the host itself. Ehrlich concluded that the drug must have a greater affinity to the parasite than to human cells. Today, the specificity of drug action is understood in terms of the drug target. An ideal target is a protein that is essential for the proliferation of the pathogen but absent in human cells. Examples are the enzymes of folate synthesis or of the nonmevalonate pathway in the malaria parasites. However, there are other ways how a drug can kill selectively. Of particular relevance is the specific activation of a prodrug inside the pathogen but not in the host, as this is how the current frontrunners of parasite chemotherapy work. Artemisinins for malaria, fexinidazole for human African trypanosomiasis, benznidazole for Chagas' disease, metronidazole for intestinal protozoa: these molecules are "magic bombs" that are triggered selectively. They are prodrugs that need to be activated by chemical reduction, i.e., the acquisition of an electron, which occurs in the parasite. Such a mode of action is shared by the novel antimalarial peroxides arterolane and artefenomel, which are activated by reduction of the endoperoxide bond with ferrous heme as the likely electron donor, a metabolic end-product of Plasmodium falciparum. Here we provide an overview on the molecular basis of selectivity of antiparasitic drug action with particular reference to the ozonides, the new generation of antimalarial peroxides designed by Jonathan Vennerstrom.
Collapse
Affiliation(s)
- Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
- University of Basel, 4001 Basel, Switzerland
| |
Collapse
|
9
|
Woodley CM, Amado PSM, Cristiano MLS, O'Neill PM. Artemisinin inspired synthetic endoperoxide drug candidates: Design, synthesis, and mechanism of action studies. Med Res Rev 2021; 41:3062-3095. [PMID: 34355414 DOI: 10.1002/med.21849] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/15/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022]
Abstract
Artemisinin combination therapies (ACTs) have been used as the first-line treatments against Plasmodium falciparum malaria for decades. Recent advances in chemical proteomics have shed light on the complex mechanism of action of semi-synthetic artemisinin (ARTs), particularly their promiscuous alkylation of parasite proteins via previous heme-mediated bioactivation of the endoperoxide bond. Alarmingly, the rise of resistance to ART in South East Asia and the synthetic limitations of the ART scaffold have pushed the course for the necessity of fully synthetic endoperoxide-based antimalarials. Several classes of synthetic endoperoxide antimalarials have been described in literature utilizing various endoperoxide warheads including 1,2-dioxanes, 1,2,4-trioxanes, 1,2,4-trioxolanes, and 1,2,4,5-tetraoxanes. Two of these classes, the 1,2,4-trioxolanes (arterolane and artefenomel) and the 1,2,4,5-tetraoxanes (N205 and E209) based antimalarials, have been explored extensively and are still in active development. In contrast, the most recent publication pertaining to the development of the 1,2-dioxane, Arteflene, and 1,2,4-trioxanes fenozan-50F, DU1301, and PA1103/SAR116242 was published in 2008. This review summarizes the synthesis, biological and clinical evaluation, and mechanistic studies of the most developed synthetic endoperoxide antimalarials, providing an update on those classes still in active development.
Collapse
Affiliation(s)
| | - Patrícia S M Amado
- Department of Chemistry, University of Liverpool, Liverpool, UK.,Center of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal.,Department of Chemistry and Pharmacy, Faculdade de Ciências e Tecnologia, University of Algarve, Faro, Portugal
| | - Maria L S Cristiano
- Center of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal.,Department of Chemistry and Pharmacy, Faculdade de Ciências e Tecnologia, University of Algarve, Faro, Portugal
| | - Paul M O'Neill
- Department of Chemistry, University of Liverpool, Liverpool, UK
| |
Collapse
|
10
|
Tisnerat C, Dassonville-Klimpt A, Gosselet F, Sonnet P. Antimalarial drug discovery: from quinine to the most recent promising clinical drug candidates. Curr Med Chem 2021; 29:3326-3365. [PMID: 34344287 DOI: 10.2174/0929867328666210803152419] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022]
Abstract
Malaria is a tropical threatening disease caused by Plasmodium parasites, resulting in 409,000 deaths in 2019. The delay of mortality and morbidity has been compounded by the widespread of drug resistant parasites from Southeast Asia since two decades. The emergence of artemisinin-resistant Plasmodium in Africa, where most cases are accounted, highlights the urgent need for new medicines. In this effort, the World Health Organization and Medicines for Malaria Venture joined to define clear goals for novel therapies and characterized the target candidate profile. This ongoing search for new treatments is based on imperative labor in medicinal chemistry which is summarized here with particular attention to hit-to-lead optimizations, key properties, and modes of action of these novel antimalarial drugs. This review, after presenting the current antimalarial chemotherapy, from quinine to the latest marketed drugs, focuses in particular on recent advances of the most promising antimalarial candidates in clinical and preclinical phases.
Collapse
Affiliation(s)
- Camille Tisnerat
- AGIR UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens. France
| | | | | | - Pascal Sonnet
- AGIR UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens. France
| |
Collapse
|
11
|
A brief history of carbon monoxide and its therapeutic origins. Nitric Oxide 2021; 111-112:45-63. [PMID: 33838343 DOI: 10.1016/j.niox.2021.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/03/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023]
Abstract
It is estimated that 10% of carbon throughout the cosmos is in the form of carbon monoxide (CO). Earth's earliest prebiotic atmosphere included the trinity of gasotransmitters CO, nitric oxide (NO), and hydrogen sulfide (H2S), for which all of life has co-evolved with. The history of CO can be loosely traced to mythological and prehistoric origins with rudimentary understanding emerging in the middle ages. Ancient literature is focused on CO's deadly toxicity which is understandable in the context of our primitive relationship with coal and fire. Scientific inquiry into CO appears to have emerged throughout the 1700s followed by chemical and toxicological profiling throughout the 1800s. Despite CO's ghastly reputation, several of the 18th and 19th century scientists suggested a therapeutic application of CO. Since 2000, the fundamental understanding of CO as a deadly nuisance has undergone a paradigm shift such that CO is now recognized as a neurotransmitter and viable pharmaceutical candidate. This review is intended to provide a brief history on the trace origins pertaining to endogenous formation and therapeutic application of CO.
Collapse
|
12
|
Artemisinin-Based Drugs Target the Plasmodium falciparum Heme Detoxification Pathway. Antimicrob Agents Chemother 2021; 65:AAC.02137-20. [PMID: 33495226 DOI: 10.1128/aac.02137-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/16/2021] [Indexed: 12/18/2022] Open
Abstract
Artemisinin (ART)-based antimalarial drugs are believed to exert lethal effects on malarial parasites by alkylating a variety of intracellular molecular targets. Recent work with live parasites has shown that one of the alkylated targets is free heme within the parasite digestive vacuole, which is liberated upon hemoglobin catabolism by the intraerythrocytic parasite, and that reduced levels of heme alkylation occur in artemisinin-resistant parasites. One implication of heme alkylation is that these drugs may inhibit parasite detoxification of free heme via inhibition of heme-to-hemozoin crystallization; however, previous reports that have investigated this hypothesis present conflicting data. By controlling reducing conditions and, hence, the availability of ferrous versus ferric forms of free heme, we modify a previously reported hemozoin inhibition assay to quantify the ability of ART-based drugs to target the heme detoxification pathway under reduced versus oxidizing conditions. Contrary to some previous reports, we find that artemisinins are potent inhibitors of hemozoin crystallization, with effective half-maximal concentrations approximately an order of magnitude lower than those for most quinoline-based antimalarial drugs. We also examine hemozoin and in vitro parasite growth inhibition for drug pairs found in the most commonly used ART-based combination therapies (ACTs). All ACTs examined inhibit hemozoin crystallization in an additive fashion, and all but one inhibit parasite growth in an additive fashion.
Collapse
|
13
|
Deda DK, Iglesias BA, Alves E, Araki K, Garcia CRS. Porphyrin Derivative Nanoformulations for Therapy and Antiparasitic Agents. Molecules 2020; 25:molecules25092080. [PMID: 32365664 PMCID: PMC7249045 DOI: 10.3390/molecules25092080] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Porphyrins and analogous macrocycles exhibit interesting photochemical, catalytic, and luminescence properties demonstrating high potential in the treatment of several diseases. Among them can be highlighted the possibility of application in photodynamic therapy and antimicrobial/antiparasitic PDT, for example, of malaria parasite. However, the low efficiency generally associated with their low solubility in water and bioavailability have precluded biomedical applications. Nanotechnology can provide efficient strategies to enhance bioavailability and incorporate targeted delivery properties to conventional pharmaceuticals, enhancing the effectiveness and reducing the toxicity, thus improving the adhesion to the treatment. In this way, those limitations can be overcome by using two main strategies: (1) Incorporation of hydrophilic substituents into the macrocycle ring while controlling the interaction with biological systems and (2) by including them in nanocarriers and delivery nanosystems. This review will focus on antiparasitic drugs based on porphyrin derivatives developed according to these two strategies, considering their vast and increasing applications befitting the multiple roles of these compounds in nature.
Collapse
Affiliation(s)
- Daiana K. Deda
- Department of Fundamental Chemistry, Institute of Chemistry, University of Sao Paulo, Av. Prof. Lineu Prestes 748, Butanta, Sao Paulo, SP 05508-000, Brazil; (D.K.D.); (K.A.)
| | - Bernardo A. Iglesias
- Bioinorganic and Porphyrinoid Materials Laboratory, Department of Chemistry, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS 97105-900, Brazil;
| | - Eduardo Alves
- Department of Life Science, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK;
| | - Koiti Araki
- Department of Fundamental Chemistry, Institute of Chemistry, University of Sao Paulo, Av. Prof. Lineu Prestes 748, Butanta, Sao Paulo, SP 05508-000, Brazil; (D.K.D.); (K.A.)
| | - Celia R. S. Garcia
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580, Sao Paulo, SP 05508-900, Brazil
- Correspondence: ; Tel.: +55-11-2648-0954
| |
Collapse
|
14
|
Secrieru A, Costa ICC, O’Neill PM, Cristiano MLS. Antimalarial Agents as Therapeutic Tools Against Toxoplasmosis-A Short Bridge between Two Distant Illnesses. Molecules 2020; 25:E1574. [PMID: 32235463 PMCID: PMC7181032 DOI: 10.3390/molecules25071574] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 11/16/2022] Open
Abstract
Toxoplasmosis is an infectious disease with paramount impact worldwide, affecting many vulnerable populations and representing a significant matter of concern. Current therapies used against toxoplasmosis are based essentially on old chemotypes, which fail in providing a definitive cure for the disease, placing the most sensitive populations at risk for irreversible damage in vital organs, culminating in death in the most serious cases. Antimalarial drugs have been shown to possess key features for drug repurposing, finding application in the treatment of other parasite-borne illnesses, including toxoplasmosis. Antimalarials provide the most effective therapeutic solutions against toxoplasmosis and make up for the majority of currently available antitoxoplasmic drugs. Additionally, other antiplasmodial drugs have been scrutinized and many promising candidates have emanated in recent developments. Available data demonstrate that it is worthwhile to explore the activity of classical and most recent antimalarial chemotypes, such as quinolines, endoperoxides, pyrazolo[1,5-a]pyrimidines, and nature-derived peptide-based parasiticidal agents, in the context of toxoplasmosis chemotherapy, in the quest for encountering more effective and safer tools for toxoplasmosis control or eradication.
Collapse
Affiliation(s)
- Alina Secrieru
- Center of Marine Sciences, CCMAR, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal; (A.S.); (I.C.C.C.)
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, FCT, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK;
| | - Inês C. C. Costa
- Center of Marine Sciences, CCMAR, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal; (A.S.); (I.C.C.C.)
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, FCT, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal
| | - Paul M. O’Neill
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK;
| | - Maria L. S. Cristiano
- Center of Marine Sciences, CCMAR, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal; (A.S.); (I.C.C.C.)
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, FCT, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal
| |
Collapse
|
15
|
Wu J, Wang X, Chiu FCK, Häberli C, Shackleford DM, Ryan E, Kamaraj S, Bulbule VJ, Wallick AI, Dong Y, White KL, Davis PH, Charman SA, Keiser J, Vennerstrom JL. Structure-Activity Relationship of Antischistosomal Ozonide Carboxylic Acids. J Med Chem 2020; 63:3723-3736. [PMID: 32134263 DOI: 10.1021/acs.jmedchem.0c00069] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Semisynthetic artemisinins and other bioactive peroxides are best known for their powerful antimalarial activities, and they also show substantial activity against schistosomes-another hemoglobin-degrading pathogen. Building on this discovery, we now describe the initial structure-activity relationship (SAR) of antischistosomal ozonide carboxylic acids OZ418 (2) and OZ165 (3). Irrespective of lipophilicity, these ozonide weak acids have relatively low aqueous solubilities and high protein binding values. Ozonides with para-substituted carboxymethoxy and N-benzylglycine substituents had high antischistosomal efficacies. It was possible to increase solubility, decrease protein binding, and maintain the high antischistosomal activity in mice infected with juvenile and adult Schistosoma mansoni by incorporating a weak base functional group in these compounds. In some cases, adding polar functional groups and heteroatoms to the spiroadamantane substructure increased the solubility and metabolic stability, but in all cases decreased the antischistosomal activity.
Collapse
Affiliation(s)
- Jianbo Wu
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States
| | - Xiaofang Wang
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States
| | - Francis C K Chiu
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Cécile Häberli
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland.,University of Basel, CH-4003 Basel, Switzerland
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Eileen Ryan
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Sriraghavan Kamaraj
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States
| | - Vivek J Bulbule
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States
| | - Alexander I Wallick
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska 68182, United States
| | - Yuxiang Dong
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States
| | - Karen L White
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska 68182, United States
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland.,University of Basel, CH-4003 Basel, Switzerland
| | - Jonathan L Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States
| |
Collapse
|