101
|
Kouakou YI, Tod M, Leboucher G, Lavoignat A, Bonnot G, Bienvenu AL, Picot S. Systematic review of artesunate pharmacokinetics: Implication for treatment of resistant malaria. Int J Infect Dis 2019; 89:30-44. [PMID: 31491558 DOI: 10.1016/j.ijid.2019.08.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Artesunate (ART) is an artemisinin derivative used as monotherapy for the treatment of severe malaria and in combination with a partner drug for non-severe malaria. Resistance of malaria parasites to artemisinins have emerged in Southeast Asia. Adjustment of drug regimen may be an option to prevent therapeutic failures considering the relative favourable safety profile of ART high doses. METHODS For that purpose, a systematic review was done using PubMed, Scopus and Web of Science databases. All studies on ART and DHA pharmacokinetic post-administration of artesunate in human patients or volunteers were included. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist 2009 was used. FINDINGS Fifty studies exploring oral, intravenous, rectal, and intramuscular route (1470 persons, volunteers and patients) were included. Correlations between artesunate doses and Cmax or AUC0-∞ of dihydroartemisinin (DHA) and DHA+ART were evaluated. This correlation was good (R2>0.9) using intravenous (IV) route. DHA and ART+DHA average concentrations (Cav) were well above estimated in vivo half-maximal effective concentration (EC50) for intravenous route, but this was not the case for oral route. INTERPRETATION The favorable Cav/EC50 ratio for IV route provides evidence that IV ART will remain efficient even in the case of increased resistance level, whereas for the oral route, a two-fold increase in EC50 may lead to therapeutic failures, thus providing a rationale for oral dose escalation. Considering the inter-individual variability of ART pharmacokinetic, Therapeutic Drug Monitoring through antimalarial stewardship activities is needed to optimize drug exposure and avoid resistance development.
Collapse
Affiliation(s)
- Yobouet Ines Kouakou
- ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Lyon University, Lyon, France
| | - Michel Tod
- Groupement Hospitalier Nord, Service Pharmacie, Hospices Civils de Lyon, Lyon, France
| | - Gilles Leboucher
- Groupement Hospitalier Nord, Service Pharmacie, Hospices Civils de Lyon, Lyon, France
| | - Adeline Lavoignat
- ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Lyon University, Lyon, France
| | - Guillaume Bonnot
- ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Lyon University, Lyon, France
| | - Anne-Lise Bienvenu
- ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Lyon University, Lyon, France; Groupement Hospitalier Nord, Service Pharmacie, Hospices Civils de Lyon, Lyon, France.
| | - Stephane Picot
- ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Lyon University, Lyon, France; Groupement Hospitalier Nord, Institut de Parasitologie et Mycologie Médicale, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
102
|
Edagha IA, Ekpo AJ, Edagha EI, Bassey JV, Nyong TP, Akpan AS, Obeten RF, Okon AS, Ating BA. Investigating the Comparative Effects of Six Artemisinin-based Combination Therapies on Plasmodium-induced Hepatorenal Toxicity. Niger Med J 2019; 60:211-218. [PMID: 31831942 PMCID: PMC6892336 DOI: 10.4103/nmj.nmj_152_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 06/30/2019] [Accepted: 07/30/2019] [Indexed: 01/23/2023] Open
Abstract
Background Too many artemisinin-based combination therapies (ACTs) are available, thus creating a dilemma on the most preferred for the treatment of malaria. Aim We compared the effect of six ACTs in mitigating Plasmodium-induced hepatorenal toxicity in experimental malaria. Materials and Methods Forty adult male Swiss mice allotted into eight groups: Group 1 (normal control [NC] uninfected and untreated), Group 2 (parasitized nontreated - [PNT]), and Groups 3-8 received Plasmodium berghei inoculum. After 72 h, the initial parasitemia was established. Groups 3-8 were administered oral therapeutic doses of artesunate-amodiaquine (AA), artesunate-mefloquine (AM), artesunate-sulfadoxine-pyrimethamine (ASP), artemisinin-piperaquine (AP), dihydroartemisinin-piperaquine (DP), and artemether-lumefantrine (AL) per kg bodyweight, respectively, as standard regimen, and final parasitemia determined. Animals were euthanized via chloroform inhalation and blood collected for hepatorenal analysis. Liver and kidney were dissected out for histology. Results Parasitemia was significantly (P < 0.05) decreased in tests compared to PNT, except in ASP group. Liver enzymes were significantly (P < 0.05) increased in PNT compared to tests and NC. Hyperplastic cells and portal tract inflammation were prominent in ASP group, but mild to moderate in other treated groups. Urea-creatinine were significantly (P < 0.05) increased in PNT compared to treated groups. The Na+ and Cl- were significantly (P < 0.05) reduced in PNT, with significantly (P < 0.05) increased K+ compared to NC and treated groups. Glomerulonephritis and glomerulus splitting was observed in PNT, while moderate distortions were observed in treated groups. The AA and AM groups had good kidney histoarchitecture. Conclusion Parasitemia decreased in all the treatment groups except in PNT and ASP groups which had severe hepatorenal distortions. Hepatorenal histoarchitecture were mildly distorted in the AA, AM and AL-administered groups with lower hepatorenal indices comparable to NC. The least elevated liver enzymes were in AA and AM. In decreasing order ASP > DP > AL > AP > AM > AA.
Collapse
Affiliation(s)
- Innocent A Edagha
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Arit J Ekpo
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Edelungudi I Edagha
- Department of Family Medicine, University of Uyo Teaching Hospital, Uyo, Nigeria
| | - Joy V Bassey
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Titus P Nyong
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Anthony S Akpan
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Rose F Obeten
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Anthony S Okon
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| | - Blessing A Ating
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Nigeria
| |
Collapse
|
103
|
Marshall JM, Raban RR, Kandul NP, Edula JR, León TM, Akbari OS. Winning the Tug-of-War Between Effector Gene Design and Pathogen Evolution in Vector Population Replacement Strategies. Front Genet 2019; 10:1072. [PMID: 31737050 PMCID: PMC6831721 DOI: 10.3389/fgene.2019.01072] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/07/2019] [Indexed: 12/19/2022] Open
Abstract
While efforts to control malaria with available tools have stagnated, and arbovirus outbreaks persist around the globe, the advent of clustered regularly interspaced short palindromic repeat (CRISPR)-based gene editing has provided exciting new opportunities for genetics-based strategies to control these diseases. In one such strategy, called "population replacement", mosquitoes, and other disease vectors are engineered with effector genes that render them unable to transmit pathogens. These effector genes can be linked to "gene drive" systems that can bias inheritance in their favor, providing novel opportunities to replace disease-susceptible vector populations with disease-refractory ones over the course of several generations. While promising for the control of vector-borne diseases on a wide scale, this sets up an evolutionary tug-of-war between the introduced effector genes and the pathogen. Here, we review the disease-refractory genes designed to date to target Plasmodium falciparum malaria transmitted by Anopheles gambiae, and arboviruses transmitted by Aedes aegypti, including dengue serotypes 2 and 3, chikungunya, and Zika viruses. We discuss resistance concerns for these effector genes, and genetic approaches to prevent parasite and viral escape variants. One general approach is to increase the evolutionary hurdle required for the pathogen to evolve resistance by attacking it at multiple sites in its genome and/or multiple stages of development. Another is to reduce the size of the pathogen population by other means, such as with vector control and antimalarial drugs. We discuss lessons learned from the evolution of resistance to antimalarial and antiviral drugs and implications for the management of resistance after its emergence. Finally, we discuss the target product profile for population replacement strategies for vector-borne disease control. This differs between early phase field trials and wide-scale disease control. In the latter case, the demands on effector gene efficacy are great; however, with new possibilities ushered in by CRISPR-based gene editing, and when combined with surveillance, monitoring, and rapid management of pathogen resistance, the odds are increasingly favoring effector genes in the upcoming evolutionary tug-of-war.
Collapse
Affiliation(s)
- John M. Marshall
- Division of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA, United States
- Innovative Genomics Institute, Berkeley, CA, United States
| | - Robyn R. Raban
- Section of Cell and Developmental Biology, University of California, San Diego, CA, United States
| | - Nikolay P. Kandul
- Section of Cell and Developmental Biology, University of California, San Diego, CA, United States
| | - Jyotheeswara R. Edula
- Section of Cell and Developmental Biology, University of California, San Diego, CA, United States
| | - Tomás M. León
- Division of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA, United States
| | - Omar S. Akbari
- Section of Cell and Developmental Biology, University of California, San Diego, CA, United States
- Tata Institute for Genetics and Society, University of California, San Diego, CA, United States
| |
Collapse
|
104
|
Negri A, Ferrari M, Nodari R, Coppa E, Mastrantonio V, Zanzani S, Porretta D, Bandi C, Urbanelli S, Epis S. Gene silencing through RNAi and antisense Vivo-Morpholino increases the efficacy of pyrethroids on larvae of Anopheles stephensi. Malar J 2019; 18:294. [PMID: 31462239 PMCID: PMC6712854 DOI: 10.1186/s12936-019-2925-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/17/2019] [Indexed: 11/25/2022] Open
Abstract
Background Insecticides are still at the core of insect pest and vector control programmes. Several lines of evidence indicate that ABC transporters are involved in detoxification processes against insecticides, including permethrin and other pyrethroids. In particular, the ABCG4 gene, a member of the G subfamily, has consistently been shown to be up-regulated in response to insecticide treatments in the mosquito malaria vector Anopheles stephensi (both adults and larvae). Methods To verify the actual involvement of this transmembrane protein in the detoxification process of permethrin, bioassays on larvae of An. stephensi, combining the insecticide with a siRNA, specifically designed for the inhibition of ABCG4 gene expression were performed. Administration to larvae of the same siRNA, labeled with a fluorescent molecule, was effected to investigate the systemic distribution of the inhibitory RNA into the larval bodies. Based on siRNA results, similar experiments using antisense Vivo-Morpholinos (Vivo-MOs) were effected. These molecules, compared to siRNA, are expected to guarantee a higher stability in environmental conditions and in the insect gut, and present thus a higher potential for future in-field applications. Results Bioassays using two different concentrations of siRNA, associated with permethrin, led to an increase of larval mortality, compared with results with permethrin alone. These outcomes confirm that ABCG4 transporter plays a role in the detoxification process against the selected insecticide. Moreover, after fluorescent labelling, it was shown the systemic dissemination of siRNA in different body districts of An. stephensi larvae, which suggest a potential systemic effect of the molecule. At the same time, results of Vivo-MO experiments were congruent with those obtained using siRNA, thus confirming the potential of ABCG4 inhibition as a strategy to increase permethrin susceptibility in mosquitoes. For the first time, Vivo-MOs were administered in water to larvae, with evidence for a biological effect. Conclusions Targeting ABCG4 gene for silencing through both techniques resulted in an increased pyrethroid efficacy. These results open the way toward the possibility to exploit ABCG4 inhibition in the context of integrated programmes for the control An. stephensi mosquitoes and malaria transmission.
Collapse
Affiliation(s)
- Agata Negri
- Department of Environmental Biology, Sapienza University of Rome, Via dei Sardi 70, 00185, Rome, Italy.,Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Via Celoria 26, 20133, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Via del Giochetto, 06126, Perugia, Italy
| | - Marco Ferrari
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Via Celoria 26, 20133, Milan, Italy.,Texas Biomedical Research Institute, San Antonio, 7620 NW Loop 410, San Antonio, TX, 78227-5301, USA
| | - Riccardo Nodari
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Via Celoria 26, 20133, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Via del Giochetto, 06126, Perugia, Italy
| | - Edoardo Coppa
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Valentina Mastrantonio
- Department of Environmental Biology, Sapienza University of Rome, Via dei Sardi 70, 00185, Rome, Italy
| | - Sergio Zanzani
- Department of Veterinary Medicine-DIMEVET, Università degli Studi di Milano, Via Celoria, 10, 20133, Milan, Italy
| | - Daniele Porretta
- Department of Environmental Biology, Sapienza University of Rome, Via dei Sardi 70, 00185, Rome, Italy
| | - Claudio Bandi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Via Celoria 26, 20133, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Via del Giochetto, 06126, Perugia, Italy
| | - Sandra Urbanelli
- Department of Environmental Biology, Sapienza University of Rome, Via dei Sardi 70, 00185, Rome, Italy
| | - Sara Epis
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Via Celoria 26, 20133, Milan, Italy. .,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Via del Giochetto, 06126, Perugia, Italy.
| |
Collapse
|
105
|
Plasmodium Genomics and Genetics: New Insights into Malaria Pathogenesis, Drug Resistance, Epidemiology, and Evolution. Clin Microbiol Rev 2019; 32:32/4/e00019-19. [PMID: 31366610 DOI: 10.1128/cmr.00019-19] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Protozoan Plasmodium parasites are the causative agents of malaria, a deadly disease that continues to afflict hundreds of millions of people every year. Infections with malaria parasites can be asymptomatic, with mild or severe symptoms, or fatal, depending on many factors such as parasite virulence and host immune status. Malaria can be treated with various drugs, with artemisinin-based combination therapies (ACTs) being the first-line choice. Recent advances in genetics and genomics of malaria parasites have contributed greatly to our understanding of parasite population dynamics, transmission, drug responses, and pathogenesis. However, knowledge gaps in parasite biology and host-parasite interactions still remain. Parasites resistant to multiple antimalarial drugs have emerged, while advanced clinical trials have shown partial efficacy for one available vaccine. Here we discuss genetic and genomic studies of Plasmodium biology, host-parasite interactions, population structures, mosquito infectivity, antigenic variation, and targets for treatment and immunization. Knowledge from these studies will advance our understanding of malaria pathogenesis, epidemiology, and evolution and will support work to discover and develop new medicines and vaccines.
Collapse
|
106
|
Zhan W, Visone J, Ouellette T, Harris JC, Wang R, Zhang H, Singh PK, Ginn J, Sukenick G, Wong TT, Okoro JI, Scales RM, Tumwebaze PK, Rosenthal PJ, Kafsack BFC, Cooper RA, Meinke PT, Kirkman LA, Lin G. Improvement of Asparagine Ethylenediamines as Anti-malarial Plasmodium-Selective Proteasome Inhibitors. J Med Chem 2019; 62:6137-6145. [PMID: 31177777 DOI: 10.1021/acs.jmedchem.9b00363] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The Plasmodium proteasome (Pf20S) emerged as a target for antimalarials. Pf20S inhibitors are active at multiple stages of the parasite life cycle and synergize with artemisinins, suggesting that Pf20S inhibitors have potential to be prophylactic, therapeutic, and transmission blocking as well as are useful for combination therapy. We recently reported asparagine ethylenediamines (AsnEDAs) as immunoproteasome inhibitors and modified AsnEDAs as selective Pf20S inhibitors. Here, we report further a structure-activity relationship study of AsnEDAs for selective inhibition of Pf20S over human proteasomes. Additionally, we show new mutation that conferred resistance to AsnEDAs and collateral sensitivity to an inhibitor of the Pf20S β2 subunit, the same as previously identified resistant mutation. This resistance could be overcome through the use of the structure-guided inhibitor design. Collateral sensitivity to inhibitors among respective proteasome subunits underscores the potential value of treating malaria with combinations of inhibitors of different proteasome subunits to minimize the emergence of drug resistance.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| | - Joseph Visone
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States.,Department of Medicine , Division of Infectious Diseases , 1300 York Avenue , New York , New York 10065 , United States
| | - Tierra Ouellette
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| | - Jacob C Harris
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States.,Department of Medicine , Division of Infectious Diseases , 1300 York Avenue , New York , New York 10065 , United States
| | - Rong Wang
- NMR Analytical Core Facility , Memorial Sloan Kettering Cancer Center , 1275 York Avenue , New York , New York 10065 , United States
| | - Hao Zhang
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| | - Pradeep K Singh
- Chemical Core Facility, Department of Biochemistry , Weill Cornell Medicine , New York , New York 10065 , United States
| | - John Ginn
- Tri-Institutional Therapeutics Discovery Institute , 413 E. 69th Street , New York , New York 10065 , United States
| | - George Sukenick
- NMR Analytical Core Facility , Memorial Sloan Kettering Cancer Center , 1275 York Avenue , New York , New York 10065 , United States
| | - Tzu-Tshin Wong
- Takeda Pharmaceutical Company Ltd. , 35 Landsdowne Street , Cambridge , Massachusetts 02139 , United States
| | - Judith I Okoro
- Infectious Diseases Research Collaboration , Kampala , Uganda
| | - Ryan M Scales
- Department of Public Health , University of North Carolina , Charlotte , North Carolina 28223 , United States
| | | | - Philip J Rosenthal
- Department of Medicine , University of California, San Francisco , San Francisco , California 94143 , United States
| | - Björn F C Kafsack
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| | - Roland A Cooper
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | - Peter T Meinke
- Tri-Institutional Therapeutics Discovery Institute , 413 E. 69th Street , New York , New York 10065 , United States
| | - Laura A Kirkman
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States.,Department of Medicine , Division of Infectious Diseases , 1300 York Avenue , New York , New York 10065 , United States
| | - Gang Lin
- Department of Microbiology & Immunology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| |
Collapse
|
107
|
Plasmodium pseudo-Tyrosine Kinase-like binds PP1 and SERA5 and is exported to host erythrocytes. Sci Rep 2019; 9:8120. [PMID: 31148576 PMCID: PMC6544628 DOI: 10.1038/s41598-019-44542-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/15/2019] [Indexed: 01/13/2023] Open
Abstract
Pseudokinases play key roles in many biological processes but they are poorly understood compared to active kinases. Eight putative pseudokinases have been predicted in Plasmodium species. We selected the unique pseudokinase belonging to tyrosine kinase like (TKL) family for detailed structural and functional analysis in P. falciparum and P. berghei. The primary structure of PfpTKL lacks residues critical for kinase activity, supporting its annotation as a pseudokinase. The recombinant pTKL pseudokinase domain was able to bind ATP, but lacked catalytic activity as predicted. The sterile alpha motif (SAM) and RVxF motifs of PfpTKL were found to interact with the P. falciparum proteins serine repeat antigen 5 (SERA5) and protein phosphatase type 1 (PP1) respectively, suggesting that pTKL has a scaffolding role. Furthermore, we found that PP1c activity in a heterologous model was modulated in an RVxF-dependent manner. During the trophozoite stages, PbpTKL was exported to infected erythrocytes where it formed complexes with proteins involved in cytoskeletal organization or host cell maturation and homeostasis. Finally, genetic analysis demonstrated that viable strains obtained by genomic deletion or knocking down PbpTKL did not affect the course of parasite intra-erythrocytic development or gametocyte emergence, indicating functional redundancy during these parasite stages.
Collapse
|
108
|
Bienvenu AL, Djimdé A, Picot S. Antimalarial stewardship programs are urgently needed for malaria elimination: a perspective. ACTA ACUST UNITED AC 2019; 26:16. [PMID: 30901307 PMCID: PMC6430615 DOI: 10.1051/parasite/2019016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/07/2019] [Indexed: 01/09/2023]
Abstract
Global malaria cases have not been significantly reduced over the last three years although more than USD 3 billion was invested in malaria control and elimination. The reasons for this stagnation are highly complex and multi-factorial. It remains that almost three billion treatment courses were supplied over the period 2010–2017: 30% of them without malaria tests, and some with suboptimal doses leading to the risk of selection of resistant parasites. An antimalarial stewardship program should be implemented at the healthcare provider, physician, pharmacist, medical student, and population levels. This would significantly reinforce the impact of international guidelines and national malaria program policies and fill the gap between recommendations and actual practices.
Collapse
Affiliation(s)
- Anne-Lise Bienvenu
- Groupement Hospitalier Nord, Service Pharmacie, Hospices Civils de Lyon, Lyon, F-69004, France - ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Université de Lyon, Villeurbanne, F-69100, France
| | - Abdoulaye Djimdé
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, PO Box: 1805 Point G, Bamako, Mali
| | - Stéphane Picot
- ICBMS CNRS 5246, SMITh, Malaria Research Unit, Campus Lyon-Tech La Doua, Université de Lyon, Villeurbanne, F-69100, France - Groupement Hospitalier Nord, Institut de Parasitologie et Mycologie Médicale, Hospices Civils de Lyon, Lyon, F-69004, France
| |
Collapse
|
109
|
Ricotta E, Kwan J. Artemisinin-Resistant Malaria as a Global Catastrophic Biological Threat. Curr Top Microbiol Immunol 2019; 424:33-57. [PMID: 31218504 DOI: 10.1007/82_2019_163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The global spread of artemisinin resistance brings with it the threat of incurable malaria. Already, this disease threatens over 219 million lives per year and causes 5-6% losses in GDP in endemic areas, even with current advances in prevention and treatment. This chapter discusses the currently tenuous position we are in globally, and the impact that could be seen if artemisinin treatment is lost, whether due to the unchecked spread of K13 mutations or poor global investment in treatment and prevention advances. Artemisinin is the backbone of current ACT treatment programs and severe malarial treatment; without it, the success of future malaria eradication programs will be in jeopardy.
Collapse
Affiliation(s)
- Emily Ricotta
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Quarters 15B-1, 8 West Dr, Bethesda, MD, 20892, USA.
- Kelly Government Solutions, Bethesda, USA.
| | - Jennifer Kwan
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Quarters 15B-1, 8 West Dr, Bethesda, MD, 20892, USA
| |
Collapse
|
110
|
Oboh MA, Ndiaye D, Antony HA, Badiane AS, Singh US, Ali NA, Bharti PK, Das A. Status of Artemisinin Resistance in Malaria Parasite Plasmodium falciparum from Molecular Analyses of the Kelch13 Gene in Southwestern Nigeria. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2305062. [PMID: 30402465 PMCID: PMC6192135 DOI: 10.1155/2018/2305062] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/20/2018] [Accepted: 09/17/2018] [Indexed: 01/24/2023]
Abstract
Evolution and spread of malaria parasite Plasmodium falciparum capable of evading antimalarials are the prime concern to malaria control. The currently effective drug, artemisinin (ART), is under threat due to detection of ART-resistant P. falciparum parasites in the Southeast Asian countries. It has been shown that amino acid (AA) mutations at the P. falciparum Kelch13 (Pfk13) gene provide resistance to ART. Nigeria, a part of the Sub-Saharan Africa, is highly endemic to malaria, contributing quite significantly to malaria, and resistance to chloroquine (CQ) and sulfadoxine-pyrimethamine (SP) combination drugs has already been reported. Since artemisinin combined therapy (ACT) is the first-line drug for treatment of uncomplicated malaria in Nigeria and five amino acid mutations have been validated in the Pfk13 gene alongside with candidate mutations for ART resistance, we performed molecular surveillance for mutations (following PCR and DNA sequence analyses) in this gene from two southwestern states of Nigeria. Statistical analyses of DNA sequences were also performed following different evolutionary models. None of the different validated and candidate AA mutations of Pfk13 gene conferring resistance to ART could be detected in P. falciparum sampled in the two southwestern states of Nigeria. In addition, DNA sequencing and sequence analyses indicated neither evolutionary selection pressure on the Pfk13 gene nor association of mutations in Pfk13 gene with mutations of other three genes conferring resistance to CQ and SP. Therefore, based on the monomorphism at the Pfk13 gene and nonassociation of mutations of this gene with mutations in three other drug-resistant genes in malaria parasite P. falciparum, it can be proposed that malaria public health is not under immediate threat in southwestern Nigeria concerning ART resistance.
Collapse
Affiliation(s)
- Mary Aigbiremo Oboh
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Daouda Ndiaye
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Hiasindh Ashmi Antony
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Aida Sadikh Badiane
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Upasana Shyamsunder Singh
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Nazia Anwar Ali
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Praveen Kumar Bharti
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Aparup Das
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| |
Collapse
|
111
|
Koller R, Mombo-Ngoma G, Grobusch MP. The early preclinical and clinical development of ganaplacide (KAF156), a novel antimalarial compound. Expert Opin Investig Drugs 2018; 27:803-810. [PMID: 30223692 DOI: 10.1080/13543784.2018.1524871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Ganaplacide (previously known as KAF156) is a novel antimalarial compound part of the imidazolopiperazine family. AREAS COVERED At the time of writing, a total of eight studies addressing its preclinical and clinical development have been published on this compound, which is currently in phase 2 of clinical development, alongside lumefantrine in a novel soluble formulation as combination partner. This review provides an overview and interpretation of the published pre-clinical and clinical data of this possible next-generation antimalarial drug. EXPERT OPINION In the search for a 'magic bullet' in malaria therapy and prophylaxis facilitating single encounter radical cure and prophylaxis, ganaplacide demonstrates some promising properties toward this ultimate goal. The available data suggest that ganaplacide exerts multi-stage antimalarial activity, and that its pharmacokinetic profile potentially allows for a simplified dosing regimen compared to that of existing antimalarial drug combinations. The first in-patient results demonstrate promising single-dose antimalarial activity, and no serious in-human safety and tolerability concerns have been reported to date.
Collapse
Affiliation(s)
- Robin Koller
- a Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases , Amsterdam University Medical Centers, University of Amsterdam , Amsterdam , The Netherlands.,b Centre de Recherches Médicales en Lambaréné (CERMEL) , Lambaréné , Gabon
| | - Ghyslain Mombo-Ngoma
- b Centre de Recherches Médicales en Lambaréné (CERMEL) , Lambaréné , Gabon.,c Institute of Tropical Medicine , University of Tübingen , Tübingen , Germany.,d Department of Tropical Medicine , Bernhard Nocht Institute for Tropical Medicine & I. Department of Medicine University Medical Center Hamburg-Eppendorf , Hamburg , Germany.,e Department of Parasitology , Université des Sciences de la Santé , Libreville , Gabon
| | - Martin P Grobusch
- a Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases , Amsterdam University Medical Centers, University of Amsterdam , Amsterdam , The Netherlands.,b Centre de Recherches Médicales en Lambaréné (CERMEL) , Lambaréné , Gabon.,c Institute of Tropical Medicine , University of Tübingen , Tübingen , Germany.,f Institute of Infectious Diseases and Molecular Medicine , University of Cape Town , Cape Town , South Africa.,g Masanga Medical Research Unit , Masanga , Sierra Leone
| |
Collapse
|
112
|
Kokkonda S, El Mazouni F, White KL, White J, Shackleford DM, Lafuente-Monasterio MJ, Rowland P, Manjalanagara K, Joseph JT, Garcia-Pérez A, Fernandez J, Gamo FJ, Waterson D, Burrows JN, Palmer MJ, Charman SA, Rathod PK, Phillips MA. Isoxazolopyrimidine-Based Inhibitors of Plasmodium falciparum Dihydroorotate Dehydrogenase with Antimalarial Activity. ACS OMEGA 2018; 3:9227-9240. [PMID: 30197997 PMCID: PMC6120730 DOI: 10.1021/acsomega.8b01573] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/31/2018] [Indexed: 06/08/2023]
Abstract
Malaria kills nearly 0.5 million people yearly and impacts the lives of those living in over 90 countries where it is endemic. The current treatment programs are threatened by increasing drug resistance. Dihydroorotate dehydrogenase (DHODH) is now clinically validated as a target for antimalarial drug discovery as a triazolopyrimidine class inhibitor (DSM265) is currently undergoing clinical development. We discovered a related isoxazolopyrimidine series in a phenotypic screen, later determining that it targeted DHODH. To determine if the isoxazolopyrimidines could yield a drug candidate, we initiated hit-to-lead medicinal chemistry. Several potent analogues were identified, including a compound that showed in vivo antimalarial activity. The isoxazolopyrimidines were more rapidly metabolized than their triazolopyrimidine counterparts, and the pharmacokinetic data were not consistent with the goal of a single-dose treatment for malaria.
Collapse
Affiliation(s)
- Sreekanth Kokkonda
- Departments
of Chemistry and Global Health, University
of Washington, Seattle, Washington 98195, United States
| | - Farah El Mazouni
- Department
of Biochemistry, University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, Texas 75390-9038, United States
| | - Karen L. White
- Centre
for Drug Candidate Optimisation, Monash
Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - John White
- Departments
of Chemistry and Global Health, University
of Washington, Seattle, Washington 98195, United States
| | - David M. Shackleford
- Centre
for Drug Candidate Optimisation, Monash
Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - Paul Rowland
- Tres
Cantos Medicines Development Campus, GSK, Severo Ochoa, Madrid 28760, Spain
| | | | | | - Adolfo Garcia-Pérez
- Tres
Cantos Medicines Development Campus, GSK, Severo Ochoa, Madrid 28760, Spain
| | - Jorge Fernandez
- Tres
Cantos Medicines Development Campus, GSK, Severo Ochoa, Madrid 28760, Spain
| | | | - David Waterson
- Medicines
for Malaria Venture, 20, Route de Pré-Bois, 1215 Geneva, Switzerland
| | - Jeremy N. Burrows
- Medicines
for Malaria Venture, 20, Route de Pré-Bois, 1215 Geneva, Switzerland
| | - Michael J. Palmer
- Medicines
for Malaria Venture, 20, Route de Pré-Bois, 1215 Geneva, Switzerland
| | - Susan A. Charman
- Centre
for Drug Candidate Optimisation, Monash
Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Pradipsinh K. Rathod
- Departments
of Chemistry and Global Health, University
of Washington, Seattle, Washington 98195, United States
| | - Margaret A. Phillips
- Department
of Biochemistry, University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, Texas 75390-9038, United States
| |
Collapse
|