1
|
Zhou Y, Wang J, Sun Y, Cheng Y, Wu W. Non-Hydroxamate Inhibitors of IspC Enzyme in the MEP Pathway: Structural Insights and Drug Development Potential. Chem Biol Drug Des 2025; 105:e70086. [PMID: 40099748 DOI: 10.1111/cbdd.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
1-Deoxy-D-xylulose-5-phosphate reductoisomerase (IspC) is a key enzyme in the MEP pathway, essential for many bacteria, human pathogens, and plants, thus being an attractive drug target. Fosmidomycin, a potent IspC inhibitor with hydroxamate metal-binding pharmacophores (MBPs), has entered clinical trials for malaria but is hampered by pharmacokinetic and toxicity issues of the hydroxamate fragment. This has led to increased interest in non-hydroxamate inhibitors. This review focuses on the crystal structure and active-site binding mode of IspC, and the structural types, inhibitory activities, and structure-activity relationships of non-hydroxamate IspC inhibitors. Early attempts to design such inhibitors involved direct removal or replacement of the hydroxamate MBPs, with varying results. Lipophilic inhibitors, bisubstrate inhibitors, and those developed for herbicidal applications have shown promise. However, challenges remain due to the sensitivity of the enzyme active site to ligand interactions. Future research could draw from other metalloenzyme studies to develop novel and efficient non-hydroxamate IspC inhibitors.
Collapse
Affiliation(s)
- Yaqing Zhou
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Jili Wang
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yong Sun
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yarui Cheng
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Wenhai Wu
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| |
Collapse
|
2
|
Diamanti E, Steinbach AM, de Carvalho LP, Ropponen HK, Lacour A, Hamid R, Eisa S, Bravo P, Bousis S, Illarionov B, Fischer M, Hamed MM, Bach NC, Rottmann M, Held J, Witschel M, Sieber SA, Hirsch AKH. Targeting the Plasmodium falciparum IspE Enzyme. ACS OMEGA 2024; 9:44465-44473. [PMID: 39524657 PMCID: PMC11541488 DOI: 10.1021/acsomega.4c06038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024]
Abstract
The enzyme IspE in Plasmodium falciparum is considered an attractive drug target, as it is essential for parasite survival and is absent in the human proteome. Yet it still has not been addressed by a small-molecule inhibitor. In this study, we conducted a high-throughput screening campaign against the PfIspE enzyme. Our approach toward a PfIspE inhibitor comprises in vitro screening, structure-activity relationship studies, examining the docking position using an AlphaFold model, and finally target verification through probe binding and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis. The newly synthesized probe containing a diazirine and an alkyne moiety (23) allowed us to demonstrate its binding to IspE in the presence of a lysate of human cells (HEK293 cells) and to get evidence that both probe 23 and the best inhibitor of the series (19) compete for the same IspE binding site.
Collapse
Affiliation(s)
- Eleonora Diamanti
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Annina M. Steinbach
- Center
for Protein Assemblies, Technical University
of Munich, Ernst-Otto-Fischer-Straße 8, 85748 Garching, Germany
| | - Lais P. de Carvalho
- Institute
of Tropical Medicine, University of Tübingen, Wilhelmstraße 27, 72074 Tübingen, Germany
| | - Henni-Karoliina Ropponen
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
- Saarland
University, Campus Building
E8.1, 66123 Saarbrücken, Germany
| | - Antoine Lacour
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
- Saarland
University, Campus Building
E8.1, 66123 Saarbrücken, Germany
| | - Rawia Hamid
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
- Saarland
University, Campus Building
E8.1, 66123 Saarbrücken, Germany
| | - Sidra Eisa
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
- Saarland
University, Campus Building
E8.1, 66123 Saarbrücken, Germany
| | - Patricia Bravo
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität
Basel Petersplatz 1, 4003 Basel, Switzerland
| | - Spyridon Bousis
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Boris Illarionov
- Hamburg
School of Food Science, Institute of Food
Chemistry, Grindelallee
117, 20146 Hamburg, Germany
| | - Markus Fischer
- Hamburg
School of Food Science, Institute of Food
Chemistry, Grindelallee
117, 20146 Hamburg, Germany
| | - Mostafa M. Hamed
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Nina C. Bach
- Center
for Protein Assemblies, Technical University
of Munich, Ernst-Otto-Fischer-Straße 8, 85748 Garching, Germany
| | - Matthias Rottmann
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität
Basel Petersplatz 1, 4003 Basel, Switzerland
| | - Jana Held
- Institute
of Tropical Medicine, University of Tübingen, Wilhelmstraße 27, 72074 Tübingen, Germany
- German
Centre for Infection Research (DZIF), 38124 Braunschweig, Germany
| | | | - Stephan A. Sieber
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
- Center
for Protein Assemblies, Technical University
of Munich, Ernst-Otto-Fischer-Straße 8, 85748 Garching, Germany
| | - Anna K. H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)-Saarland University,
Department of Pharmacy, Helmholtz Centre
for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany
- Saarland
University, Campus Building
E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
3
|
Willocx D, Bizzarri L, Alhayek A, Kannan D, Bravo P, Illarionov B, Rox K, Lohse J, Fischer M, Kany AM, Hahne H, Rottmann M, Witschel M, Odom John A, Hamed MM, Diamanti E, Hirsch AKH. Targeting Plasmodium falciparum IspD in the Methyl-d-erythritol Phosphate Pathway: Urea-Based Compounds with Nanomolar Potency on Target and Low-Micromolar Whole-Cell Activity. J Med Chem 2024; 67:17070-17086. [PMID: 39303294 PMCID: PMC11472328 DOI: 10.1021/acs.jmedchem.4c00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/26/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
The methyl-d-erythritol phosphate (MEP) pathway has emerged as an interesting target in the fight against antimicrobial resistance. The pathway is essential in many human pathogens, including Plasmodium falciparum (Pf), but is absent in human cells. In the present study, we report on the discovery of a new chemical class targeting IspD, the third enzyme in the pathway. Exploration of the structure-activity relationship yielded inhibitors with potency in the low-nanomolar range. Moreover, we investigated the whole-cell activity, mode of inhibition, metabolic, and plasma stability of this compound class, and conducted in vivo pharmacokinetic profiling on selected compounds. Lastly, we disclosed a new mass spectrometry (MS)-based enzymatic assay for direct IspD activity determination, circumventing the need for auxiliary enzymes. In summary, we have identified a readily synthesizable compound class, demonstrating excellent activity and a promising profile, positioning it as a valuable tool compound for advancing research on IspD.
Collapse
Affiliation(s)
- Daan Willocx
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Lorenzo Bizzarri
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- OmicScouts
GmbH, Lise-Meitner-Straße
30, 85354 Freising, Germany
| | - Alaa Alhayek
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Deepika Kannan
- Department
of Pediatrics, Children’s Hospital
of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Patricia Bravo
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität
Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Boris Illarionov
- Hamburg
School of Food Science, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Jonas Lohse
- OmicScouts
GmbH, Lise-Meitner-Straße
30, 85354 Freising, Germany
| | - Markus Fischer
- Hamburg
School of Food Science, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany
| | - Andreas M. Kany
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Hannes Hahne
- OmicScouts
GmbH, Lise-Meitner-Straße
30, 85354 Freising, Germany
| | - Matthias Rottmann
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität
Basel, Petersplatz 1, 4003 Basel, Switzerland
| | | | - Audrey Odom John
- Department
of Pediatrics, Children’s Hospital
of Philadelphia, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Mostafa M. Hamed
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Eleonora Diamanti
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Anna K. H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
4
|
Woodland JG, Horatscheck A, Soares de Melo C, Dziwornu GA, Taylor D. Another decade of antimalarial drug discovery: New targets, tools and molecules. PROGRESS IN MEDICINAL CHEMISTRY 2024; 63:161-234. [PMID: 39370241 DOI: 10.1016/bs.pmch.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Malaria remains a devastating but preventable infectious disease that disproportionately affects the African continent. Emerging resistance to current frontline therapies means that not only are new treatments urgently required, but also novel validated antimalarial targets to circumvent cross-resistance. Fortunately, tremendous efforts have been made by the global drug discovery community over the past decade. In this chapter, we will highlight some of the antimalarial drug discovery and development programmes currently underway across the globe, charting progress in the identification of new targets and the development of new classes of drugs to prosecute them. These efforts have been complemented by the development of valuable tools to accelerate target validation such as the NOD scid gamma (NSG) humanized mouse efficacy model and progress in predictive modelling and open-source software. Among the medicinal chemistry programmes that have been conducted over the past decade are those targeting Plasmodium falciparum ATPase4 (ATP4) and acetyl-CoA synthetase (AcAS) as well as proteins disrupting parasite protein translation such as the aminoacyl-tRNA synthetases (aaRSs) and eukaryotic elongation factor 2 (eEF2). The benefits and challenges of targeting Plasmodium kinases will be examined, with a focus on Plasmodium cyclic GMP-dependent protein kinase (PKG), cyclin-dependent-like protein kinase 3 (CLK3) and phosphatidylinositol 4-kinase (PI4K). The chapter concludes with a survey of incipient drug discovery centres in Africa and acknowledges the value of recent international meetings in galvanizing and uniting the antimalarial drug discovery community.
Collapse
Affiliation(s)
- John G Woodland
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - André Horatscheck
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
| | - Candice Soares de Melo
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
| | - Godwin A Dziwornu
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
| | - Dale Taylor
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa.
| |
Collapse
|
5
|
Chen X, Zhao H, Wang C, Hamed M, Shang Q, Yang Y, Diao X, Sun X, Hu W, Jiang X, Zhang Y, Hirsch AKH, Wu D, Zhuang J. Two natural compounds as potential inhibitors against the Helicobacter pylori and Acinetobacter baumannii IspD enzymes. Int J Antimicrob Agents 2024; 63:107160. [PMID: 38537721 DOI: 10.1016/j.ijantimicag.2024.107160] [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: 12/30/2023] [Revised: 02/27/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
In a vast majority of bacteria, protozoa and plants, the methylerythritol phosphate (MEP) pathway is utilized for the synthesis of isopentenyl diphosphate (IDP) and dimethylallyl diphosphate (DMADP), which are precursors for isoprenoids. Isoprenoids, such as cholesterol and coenzyme Q, play a variety of crucial roles in physiological activities, including cell-membrane formation, protein degradation, cell apoptosis, and transcription regulation. In contrast, humans employ the mevalonate (MVA) pathway for the production of IDP and DMADP, rendering proteins in the MEP pathway appealing targets for antimicrobial agents. This pathway consists of seven consecutive enzymatic reactions, of which 4-diphosphocytidyl-2C-methyl-D-erythritol synthase (IspD) and 2C-methyl-D-erythritol 2,4-cyclodiphosphate synthase (IspF) catalyze the third and fifth steps, respectively. In this study, we characterized the enzymatic activities and protein structures of Helicobacter pylori IspDF and Acinetobacter baumannii IspD. Then, using the direct interaction-based thermal shift assay, we conducted a compound screening of an approved drug library and identified 27 hit compounds potentially binding to AbIspD. Among them, two natural products, rosmarinic acid and tanshinone IIA sodium sulfonate, exhibited inhibitory activities against HpIspDF and AbIspD, by competing with one of the substrates, MEP. Moreover, tanshinone IIA sodium sulfonate also demonstrated certain antibacterial effects against H. pylori. In summary, we identified two IspD inhibitors from approved ingredients, broadening the scope for antibiotic discovery targeting the MEP pathway.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Huilin Zhao
- Department of Pathogenic Biology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Chuandong Wang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Mostafa Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbrücken, Germany
| | - Qinghong Shang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Yating Yang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiaotong Diao
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiangnan Sun
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Wei Hu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xukai Jiang
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| | - Youming Zhang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Dalei Wu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.
| | - Jingjing Zhuang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China; Marine College, Shandong University, Weihai, China.
| |
Collapse
|
6
|
Wu X, Bu M, Yang Z, Ping H, Song C, Duan J, Zhang A. Design and synthesis of fosmidomycin analogs containing aza-linkers and their biological activity evaluation. PEST MANAGEMENT SCIENCE 2024; 80:846-856. [PMID: 37794283 DOI: 10.1002/ps.7810] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND The enzymes involved in the 2-C-methyl-d-erythritol 4-phosphate (MEP) pathway are attractive targets of a new mode of action for developing anti-infective drugs and herbicides, and inhibitors against 1-deoxy-d-xylulose 5-phosphate reductoisomerase (IspC), the second key enzyme in the pathway, have been intensively investigated; however, few works are reported regarding IspC inhibitors designed for new herbicide discovery. RESULTS A series of fosmidomycin (FOS) analogs were designed with nitrogen-containing linkers replacing the trimethylene linker between the two active substructures of FOS, phosphonic acid and hydroxamic acid. Synthesis followed a facile three-step route of sequential aza-Michael addition of α-amino acids to dibenzyl vinylphosphonate, amidation of the amino acid carboxyl with O-benzyl hydroxylamine, and simultaneous removal of the benzyl protective groups. Biological activity evaluation of IspC and model plants revealed that some compounds had moderate enzyme and model plant growth inhibition effects. In particular, compound 10g, which has a N-(4-fluorophenylethyl) nitrogen-containing linker, exhibited the best plant inhibition activities, superior to the control FOS against the model plants Arabidopsis thaliana, Brassica napus L., Amaranthus retroflexus and Echinochloa crus-galli. A dimethylallyl pyrophosphate rescue assay on A. thaliana confirmed that both 10g and FOS exert their herbicidal activity by blocking the MEP pathway. This result consistent with molecular docking, which confirmed 10g and FOS binding to the IspC active site in a similar way. CONCLUSION Compound 10g has excellent herbicidal activity and represents the first herbicide lead structure of a new mode of action that targets IspC enzyme in the MEP pathway. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xin Wu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Mengwei Bu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Zili Yang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Hongrui Ping
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Chunlin Song
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jiang Duan
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Aidong Zhang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| |
Collapse
|
7
|
Quansah N, Sarah C, Yamaryo-Botté Y, Botté CY. Complex Endosymbiosis II: The Nonphotosynthetic Plastid of Apicomplexa Parasites (The Apicoplast) and Its Integrated Metabolism. Methods Mol Biol 2024; 2776:43-62. [PMID: 38502497 DOI: 10.1007/978-1-0716-3726-5_3] [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: 03/21/2024]
Abstract
Chloroplasts are essential organelles that are responsible for photosynthesis in a wide range of organisms that have colonized all biotopes on Earth such as plants and unicellular algae. Interestingly, a secondary endosymbiotic event of a red algal ancestor gave rise to a group of organisms that have adopted an obligate parasitic lifestyle named Apicomplexa parasites. Apicomplexa parasites are some of the most widespread and poorly controlled pathogens in the world. These infectious agents are responsible for major human diseases such as toxoplasmosis, caused by Toxoplasma gondii, and malaria, caused by Plasmodium spp. Most of these parasites harbor this relict plastid named the apicoplast, which is essential for parasite survival. The apicoplast has lost photosynthetic capacities but is metabolically similar to plant and algal chloroplasts. The apicoplast is considered a novel and important drug target against Apicomplexa parasites. This chapter focuses on the apicoplast of apicomplexa parasites, its maintenance, and its metabolic pathways.
Collapse
Affiliation(s)
- Nyamekye Quansah
- ApicoLipid Team, Institute for Advanced Biosciences, UMR5309, Centre National de la Recherche Scientifique, Université Grenoble Alpes, U1209, Institut National de la Santé et de la Recherche Médicale, Grenoble, France
| | - Charital Sarah
- ApicoLipid Team, Institute for Advanced Biosciences, UMR5309, Centre National de la Recherche Scientifique, Université Grenoble Alpes, U1209, Institut National de la Santé et de la Recherche Médicale, Grenoble, France
| | - Yoshiki Yamaryo-Botté
- ApicoLipid Team, Institute for Advanced Biosciences, UMR5309, Centre National de la Recherche Scientifique, Université Grenoble Alpes, U1209, Institut National de la Santé et de la Recherche Médicale, Grenoble, France
| | - Cyrille Y Botté
- ApicoLipid Team, Institute for Advanced Biosciences, UMR5309, Centre National de la Recherche Scientifique, Université Grenoble Alpes, U1209, Institut National de la Santé et de la Recherche Médicale, Grenoble, France.
- Centre National de la Recherche Scientifique, Institute for Advanced Biosciences, UMR5309, Université Grenoble Alpes, INSERM, U1209, Grenoble, France.
| |
Collapse
|
8
|
Crispim M, Verdaguer IB, Hernández A, Kronenberger T, Fenollar À, Yamaguchi LF, Alberione MP, Ramirez M, de Oliveira SS, Katzin AM, Izquierdo L. Beyond the MEP Pathway: A novel kinase required for prenol utilization by malaria parasites. PLoS Pathog 2024; 20:e1011557. [PMID: 38277417 PMCID: PMC10849223 DOI: 10.1371/journal.ppat.1011557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/07/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
A proposed treatment for malaria is a combination of fosmidomycin and clindamycin. Both compounds inhibit the methylerythritol 4-phosphate (MEP) pathway, the parasitic source of farnesyl and geranylgeranyl pyrophosphate (FPP and GGPP, respectively). Both FPP and GGPP are crucial for the biosynthesis of several essential metabolites such as ubiquinone and dolichol, as well as for protein prenylation. Dietary prenols, such as farnesol (FOH) and geranylgeraniol (GGOH), can rescue parasites from MEP inhibitors, suggesting the existence of a missing pathway for prenol salvage via phosphorylation. In this study, we identified a gene in the genome of P. falciparum, encoding a transmembrane prenol kinase (PolK) involved in the salvage of FOH and GGOH. The enzyme was expressed in Saccharomyces cerevisiae, and its FOH/GGOH kinase activities were experimentally validated. Furthermore, conditional knockout parasites (Δ-PolK) were created to investigate the biological importance of the FOH/GGOH salvage pathway. Δ-PolK parasites were viable but displayed increased susceptibility to fosmidomycin. Their sensitivity to MEP inhibitors could not be rescued by adding prenols. Additionally, Δ-PolK parasites lost their capability to utilize prenols for protein prenylation. Experiments using culture medium supplemented with whole/delipidated human plasma in transgenic parasites revealed that human plasma has components that can diminish the effectiveness of fosmidomycin. Mass spectrometry tests indicated that both bovine supplements used in culture and human plasma contain GGOH. These findings suggest that the FOH/GGOH salvage pathway might offer an alternate source of isoprenoids for malaria parasites when de novo biosynthesis is inhibited. This study also identifies a novel kind of enzyme related to isoprenoid metabolism.
Collapse
Affiliation(s)
- Marcell Crispim
- Department of Parasitology, Institute of Biomedical Sciences of the University of São Paulo, São Paulo, Brazil
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Ignasi Bofill Verdaguer
- Department of Parasitology, Institute of Biomedical Sciences of the University of São Paulo, São Paulo, Brazil
| | - Agustín Hernández
- Center for Biological and Health Sciences, Integrated Unit for Research in Biodiversity (BIOTROP-CCBS), Federal University of São Carlos, São Carlos, Brazil
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany
| | - Àngel Fenollar
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | | | - María Pía Alberione
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Miriam Ramirez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | | | - Alejandro Miguel Katzin
- Department of Parasitology, Institute of Biomedical Sciences of the University of São Paulo, São Paulo, Brazil
| | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
9
|
Ropponen HK, Diamanti E, Johannsen S, Illarionov B, Hamid R, Jaki M, Sass P, Fischer M, Haupenthal J, Hirsch AKH. Exploring the Translational Gap of a Novel Class of Escherichia coli IspE Inhibitors. ChemMedChem 2023; 18:e202300346. [PMID: 37718320 DOI: 10.1002/cmdc.202300346] [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: 07/05/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 09/19/2023]
Abstract
Discovery of novel antibiotics needs multidisciplinary approaches to gain target enzyme and bacterial activities while aiming for selectivity over mammalian cells. Here, we report a multiparameter optimisation of a fragment-like hit that was identified through a structure-based virtual-screening campaign on Escherichia coli IspE crystal structure. Subsequent medicinal-chemistry design resulted in a novel class of E. coli IspE inhibitors, exhibiting activity also against the more pathogenic bacteria Pseudomonas aeruginosa and Acinetobacter baumannii. While cytotoxicity remains a challenge for the series, it provides new insights on the molecular properties for balancing enzymatic target and bacterial activities simultaneously as well as new starting points for the development of IspE inhibitors with a predicted new mode of action.
Collapse
Affiliation(s)
- Henni-Karoliina Ropponen
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
- Current address: AMR Action Fund GP GmbH, Messeplatz 10, 4058, Basel, Switzerland
| | - Eleonora Diamanti
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Sandra Johannsen
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Boris Illarionov
- Hamburg School of Food Science, Institute of Food Chemistry, Grindelallee 117, 20146, Hamburg, Germany
| | - Rawia Hamid
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Miriam Jaki
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
- Current address: University of Freiburg, Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Sonnenstraße 5, 79104, Freiburg, Germany
| | - Peter Sass
- Interfaculty Institute of Microbiology and Infection Medicine, Universität Tubingen
| | - Markus Fischer
- Hamburg School of Food Science, Institute of Food Chemistry, Grindelallee 117, 20146, Hamburg, Germany
| | - Jörg Haupenthal
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Anna K H Hirsch
- Drug Discovery and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| |
Collapse
|
10
|
Nguyen TD, Gao B, Amaratunga C, Dhorda M, Tran TNA, White NJ, Dondorp AM, Boni MF, Aguas R. Preventing antimalarial drug resistance with triple artemisinin-based combination therapies. Nat Commun 2023; 14:4568. [PMID: 37516752 PMCID: PMC10387089 DOI: 10.1038/s41467-023-39914-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 07/04/2023] [Indexed: 07/31/2023] Open
Abstract
Increasing levels of artemisinin and partner drug resistance threaten malaria control and elimination globally. Triple artemisinin-based combination therapies (TACTs) which combine artemisinin derivatives with two partner drugs are efficacious and well tolerated in clinical trials, including in areas of multidrug-resistant malaria. Whether early TACT adoption could delay the emergence and spread of antimalarial drug resistance is a question of vital importance. Using two independent individual-based models of Plasmodium falciparum epidemiology and evolution, we evaluated whether introduction of either artesunate-mefloquine-piperaquine or artemether-lumefantrine-amodiaquine resulted in lower long-term artemisinin-resistance levels and treatment failure rates compared with continued ACT use. We show that introduction of TACTs could significantly delay the emergence and spread of artemisinin resistance and treatment failure, extending the useful therapeutic life of current antimalarial drugs, and improving the chances of malaria elimination. We conclude that immediate introduction of TACTs should be considered by policy makers in areas of emerging artemisinin resistance.
Collapse
Affiliation(s)
- Tran Dang Nguyen
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA
| | - Bo Gao
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chanaki Amaratunga
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Mehul Dhorda
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thu Nguyen-Anh Tran
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA
| | - Nicholas J White
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arjen M Dondorp
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Maciej F Boni
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, USA.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Ricardo Aguas
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
11
|
Recent approaches in the drug research and development of novel antimalarial drugs with new targets. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:1-27. [PMID: 36692468 DOI: 10.2478/acph-2023-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 01/25/2023]
Abstract
Malaria is a serious worldwide medical issue that results in substantial annual death and morbidity. The availability of treatment alternatives is limited, and the rise of resistant parasite types has posed a significant challenge to malaria treatment. To prevent a public health disaster, novel antimalarial agents with single-dosage therapies, extensive curative capability, and new mechanisms are urgently needed. There are several approaches to developing antimalarial drugs, ranging from alterations of current drugs to the creation of new compounds with specific targeting abilities. The availability of multiple genomic techniques, as well as recent advancements in parasite biology, provides a varied collection of possible targets for the development of novel treatments. A number of promising pharmacological interference targets have been uncovered in modern times. As a result, our review concentrates on the most current scientific and technical progress in the innovation of new antimalarial medications. The protein kinases, choline transport inhibitors, dihydroorotate dehydrogenase inhibitors, isoprenoid biosynthesis inhibitors, and enzymes involved in the metabolism of lipids and replication of deoxyribonucleic acid, are among the most fascinating antimalarial target proteins presently being investigated. The new cellular targets and drugs which can inhibit malaria and their development techniques are summarised in this study.
Collapse
|
12
|
Harbin A, Laventhal N, Navin M. Ethics of age de-escalation in pediatric vaccine trials: Attending to the case of COVID-19. Vaccine 2023; 41:1584-1588. [PMID: 36732168 PMCID: PMC9888531 DOI: 10.1016/j.vaccine.2023.01.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/28/2023]
Abstract
In the development of new vaccines, many trials use age de-escalation: after establishing safety and efficacy in adult populations, progressively younger cohorts are enrolled and studied. Age de-escalation promotes many values. The responsibility to protect children from potential risks of experimental vaccines is significant, not only given increased risks of adverse effects but also because parents and medical professionals have a moral responsibility to protect children from harms associated with novel, uncertain interventions. Further, given that young children cannot provide informed consent, acceptable risks for research requiring proxy consent are lower than for adults making decisions for themselves. Although age de-escalation approaches are widely used in vaccine trials, including notably in the recent development of pediatric COVID-19 vaccines, ethicists have not addressed the benefits and risks of these approaches. Their benefits are largely assumed and unstated, while their potential risks are usually overlooked. There are no official ethics guidelines for the use of age de-escalation in clinical research. In this paper, we provide a systematic account of key moral factors to consider when employing age de-escalation. Analyzing pediatric COVID-19 vaccine development as our key case study, we clarify the benefits, risks, and trade-offs involved in age de-escalation approaches and call for the development of evidence-based best practice guidelines to identify when age de-escalation is likely to be an ethical strategy in vaccine development.
Collapse
Affiliation(s)
- Ami Harbin
- Department of Philosophy, Oakland University, Rochester, MI, USA.
| | - Naomi Laventhal
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Center for Bioethics and Social Sciences in Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mark Navin
- Department of Philosophy, Oakland University, Rochester, MI, USA; Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA; Clinical Ethics Consultation Service, Department of Spiritual Care, Corewell East, Southfield, MI, USA
| |
Collapse
|
13
|
Elahi R, Prigge ST. New insights into apicoplast metabolism in blood-stage malaria parasites. Curr Opin Microbiol 2023; 71:102255. [PMID: 36563485 PMCID: PMC9852000 DOI: 10.1016/j.mib.2022.102255] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
The apicoplast of Plasmodium falciparum is the only source of essential isoprenoid precursors and Coenzyme A (CoA) in the parasite. Isoprenoid precursor synthesis relies on the iron-sulfur cluster (FeS) cofactors produced within the apicoplast, rendering FeS synthesis an essential function of this organelle. Recent reports provide important insights into the roles of FeS cofactors and the use of isoprenoid precursors and CoA both inside and outside the apicoplast. Here, we review the recent insights into the roles of these metabolites in blood-stage malaria parasites and discuss new questions that have been raised in light of these discoveries.
Collapse
Affiliation(s)
- Rubayet Elahi
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA; The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Sean T Prigge
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA; The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA.
| |
Collapse
|
14
|
Characterization of Domiphen Bromide as a New Fast-Acting Antiplasmodial Agent Inhibiting the Apicoplastidic Methyl Erythritol Phosphate Pathway. Pharmaceutics 2022; 14:pharmaceutics14071320. [PMID: 35890216 PMCID: PMC9319574 DOI: 10.3390/pharmaceutics14071320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/17/2022] Open
Abstract
The evolution of resistance by the malaria parasite to artemisinin, the key component of the combination therapy strategies that are at the core of current antimalarial treatments, calls for the urgent identification of new fast-acting antimalarials. The apicoplast organelle is a preferred target of antimalarial drugs because it contains biochemical processes absent from the human host. Fosmidomycin is the only drug in clinical trials targeting the apicoplast, where it inhibits the methyl erythritol phosphate (MEP) pathway. Here, we characterized the antiplasmodial activity of domiphen bromide (DB), another MEP pathway inhibitor with a rapid mode of action that arrests the in vitro growth of Plasmodium falciparum at the early trophozoite stage. Metabolomic analysis of the MEP pathway and Krebs cycle intermediates in 20 µM DB-treated parasites suggested a rapid activation of glycolysis with a concomitant decrease in mitochondrial activity, consistent with a rapid killing of the pathogen. These results present DB as a model compound for the development of new, potentially interesting drugs for future antimalarial combination therapies.
Collapse
|
15
|
Diamanti E, Hamed MM, Lacour A, Bravo P, Illarionov B, Fischer M, Rottmann M, Witschel M, Hirsch AKH. Targeting the IspD Enzyme in the MEP Pathway: Identification of a Novel Fragment Class. ChemMedChem 2022; 17:e202100679. [PMID: 34918860 PMCID: PMC9305118 DOI: 10.1002/cmdc.202100679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/14/2021] [Indexed: 12/01/2022]
Abstract
The enzymes of the 2-C-methylerythritol-d-erythritol 4-phosphate (MEP) pathway (MEP pathway or non-mevalonate pathway) are responsible for the synthesis of universal precursors of the large and structurally diverse family of isoprenoids. This pathway is absent in humans, but present in many pathogenic organisms and plants, making it an attractive source of drug targets. Here, we present a high-throughput screening approach that led to the discovery of a novel fragment hit active against the third enzyme of the MEP pathway, PfIspD. A systematic SAR investigation afforded a novel chemical structure with a balanced activity-stability profile (16). Using a homology model of PfIspD, we proposed a putative binding mode for our newly identified inhibitors that sets the stage for structure-guided optimization.
Collapse
Affiliation(s)
- Eleonora Diamanti
- Helmholtz Institute for Pharmaceutical Research (HIPS)Helmholtz Centre for Infection Research (HZI)Saarland University Campus E8.166123SaarbrückenGermany
- Helmholtz International Lab for Anti-InfectivesSaarland University Campus E8.166123SaarbrückenGermany
| | - Mostafa M. Hamed
- Helmholtz Institute for Pharmaceutical Research (HIPS)Helmholtz Centre for Infection Research (HZI)Saarland University Campus E8.166123SaarbrückenGermany
| | - Antoine Lacour
- Helmholtz Institute for Pharmaceutical Research (HIPS)Helmholtz Centre for Infection Research (HZI)Saarland University Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| | - Patricia Bravo
- Swiss Tropical and Public Health InstituteSocinstrasse 574002BaselSwitzerland
- Universität BaselPetersplatz 14003BaselSwitzerland
| | - Boris Illarionov
- Hamburg School of Food ScienceUniversity of HamburgGrindelallee 11720146HamburgGermany
| | - Markus Fischer
- Hamburg School of Food ScienceUniversity of HamburgGrindelallee 11720146HamburgGermany
| | - Matthias Rottmann
- Swiss Tropical and Public Health InstituteSocinstrasse 574002BaselSwitzerland
- Universität BaselPetersplatz 14003BaselSwitzerland
| | | | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research (HIPS)Helmholtz Centre for Infection Research (HZI)Saarland University Campus E8.166123SaarbrückenGermany
- Helmholtz International Lab for Anti-InfectivesSaarland University Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| |
Collapse
|
16
|
Orwa TO, Mbogo RW, Luboobi LS. Optimal control analysis of hepatocytic-erythrocytic dynamics of Plasmodium falciparum malaria. Infect Dis Model 2022; 7:82-108. [PMID: 34977436 PMCID: PMC8686038 DOI: 10.1016/j.idm.2021.11.006] [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: 06/17/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
This paper presents an in-host malaria model subject to anti-malarial drug treatment and malaria vaccine antigens combinations. Pontryagin's Maximum Principle is applied to establish optimal control strategies against infected erythrocytes, infected hepatocytes and malaria parasites. Results from numerical simulation reveal that a combination of pre-erythrocytic vaccine antigen, blood schizontocide and gametocytocide drugs would offer the best strategy to eradicate clinical P. falciparum malaria. Sensitivity analysis, further reveal that the efficacy of blood schizontocides and blood stage vaccines are crucial in the control of clinical malaria infection. Futhermore, we found that an effective blood schizontocide should be used alongside efficacious blood stage vaccine for rapid eradication of infective malaria parasites. The authors hope that the results of this study will help accelerate malaria elimination efforts by combining malaria vaccines and anti-malarial drugs against the deadly P. falciparum malaria.
Collapse
Affiliation(s)
- Titus Okello Orwa
- Institute of Mathematical Sciences, Strathmore University, P.O Box 59 857-00 200, Nairobi, Kenya
| | - Rachel Waema Mbogo
- Institute of Mathematical Sciences, Strathmore University, P.O Box 59 857-00 200, Nairobi, Kenya
| | | |
Collapse
|
17
|
Jobelius H, Bianchino GI, Borel F, Chaignon P, Seemann M. The Reductive Dehydroxylation Catalyzed by IspH, a Source of Inspiration for the Development of Novel Anti-Infectives. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030708. [PMID: 35163971 PMCID: PMC8837944 DOI: 10.3390/molecules27030708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/11/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022]
Abstract
The non-mevalonate or also called MEP pathway is an essential route for the biosynthesis of isoprenoid precursors in most bacteria and in microorganisms belonging to the Apicomplexa phylum, such as the parasite responsible for malaria. The absence of this pathway in mammalians makes it an interesting target for the discovery of novel anti-infectives. As last enzyme of this pathway, IspH is an oxygen sensitive [4Fe-4S] metalloenzyme that catalyzes 2H+/2e− reductions and a water elimination by involving non-conventional bioinorganic and bioorganometallic intermediates. After a detailed description of the discovery of the [4Fe-4S] cluster of IspH, this review focuses on the IspH mechanism discussing the results that have been obtained in the last decades using an approach combining chemistry, enzymology, crystallography, spectroscopies, and docking calculations. Considering the interesting druggability of this enzyme, a section about the inhibitors of IspH discovered up to now is reported as well. The presented results constitute a useful and rational help to inaugurate the design and development of new potential chemotherapeutics against pathogenic organisms.
Collapse
Affiliation(s)
- Hannah Jobelius
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg UMR 7177, Université de Strasbourg/CNRS, 4, rue Blaise Pascal, 67070 Strasbourg, France; (H.J.); (G.I.B.); (P.C.)
| | - Gabriella Ines Bianchino
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg UMR 7177, Université de Strasbourg/CNRS, 4, rue Blaise Pascal, 67070 Strasbourg, France; (H.J.); (G.I.B.); (P.C.)
| | - Franck Borel
- Institut de Biologie Structurale, Université Grenoble Alpes/CEA/CNRS, 38000 Grenoble, France;
| | - Philippe Chaignon
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg UMR 7177, Université de Strasbourg/CNRS, 4, rue Blaise Pascal, 67070 Strasbourg, France; (H.J.); (G.I.B.); (P.C.)
| | - Myriam Seemann
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg UMR 7177, Université de Strasbourg/CNRS, 4, rue Blaise Pascal, 67070 Strasbourg, France; (H.J.); (G.I.B.); (P.C.)
- Correspondence:
| |
Collapse
|
18
|
Zhu D, Johannsen S, Masini T, Simonin C, Haupenthal J, Illarionov B, Andreas A, Awale M, Gierse RM, van der Laan T, van der Vlag R, Nasti R, Poizat M, Buhler E, Reiling N, Müller R, Fischer M, Reymond JL, Hirsch AKH. Discovery of novel drug-like antitubercular hits targeting the MEP pathway enzyme DXPS by strategic application of ligand-based virtual screening. Chem Sci 2022; 13:10686-10698. [PMID: 36320685 PMCID: PMC9491098 DOI: 10.1039/d2sc02371g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/07/2022] [Indexed: 12/04/2022] Open
Abstract
In the present manuscript, we describe how we successfully used ligand-based virtual screening (LBVS) to identify two small-molecule, drug-like hit classes with excellent ADMET profiles against the difficult to address microbial enzyme 1-deoxy-d-xylulose-5-phosphate synthase (DXPS). In the fight against antimicrobial resistance (AMR), it has become increasingly important to address novel targets such as DXPS, the first enzyme of the 2-C-methyl-d-erythritol-4-phosphate (MEP) pathway, which affords the universal isoprenoid precursors. This pathway is absent in humans but essential for pathogens such as Mycobacterium tuberculosis, making it a rich source of drug targets for the development of novel anti-infectives. Standard computer-aided drug-design tools, frequently applied in other areas of drug development, often fail for targets with large, hydrophilic binding sites such as DXPS. Therefore, we introduce the concept of pseudo-inhibitors, combining the benefits of pseudo-ligands (defining a pharmacophore) and pseudo-receptors (defining anchor points in the binding site), for providing the basis to perform a LBVS against M. tuberculosis DXPS. Starting from a diverse set of reference ligands showing weak inhibition of the orthologue from Deinococcus radiodurans DXPS, we identified three structurally unrelated classes with promising in vitro (against M. tuberculosis DXPS) and whole-cell activity including extensively drug-resistant strains of M. tuberculosis. The hits were validated to be specific inhibitors of DXPS and to have a unique mechanism of inhibition. Furthermore, two of the hits have a balanced profile in terms of metabolic and plasma stability and display a low frequency of resistance development, making them ideal starting points for hit-to-lead optimization of antibiotics with an unprecedented mode of action. We identified two drug-like antitubercular hits with submicromolar inhibition constants against the target 1-deoxy-d-xylulose-5-phosphate synthase (DXPS) with a new mode of action and promising activity against drug-resistant tuberculosis.![]()
Collapse
Affiliation(s)
- Di Zhu
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1 66123 Saarbrücken Germany
- Stratingh Institute for Chemistry, University of Groningen Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Sandra Johannsen
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1 66123 Saarbrücken Germany
| | - Tiziana Masini
- Stratingh Institute for Chemistry, University of Groningen Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Céline Simonin
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern Freiestrasse 3 3012 Bern Switzerland
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
| | - Boris Illarionov
- Hamburg School of Food Science, Institute of Food Chemistry Grindelallee 117 20146 Hamburg Germany
| | - Anastasia Andreas
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1 66123 Saarbrücken Germany
| | - Mahendra Awale
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern Freiestrasse 3 3012 Bern Switzerland
| | - Robin M Gierse
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1 66123 Saarbrücken Germany
- Stratingh Institute for Chemistry, University of Groningen Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Tridia van der Laan
- Department of Mycobacteria, National Institute of Public Health and the Environment (RIVM), Diagnostics and Laboratory Surveillance (IDS) Infectious Diseases Research Antonie van Leeuwenhoeklaan 9 3721 MA Bilthoven The Netherlands
| | - Ramon van der Vlag
- Stratingh Institute for Chemistry, University of Groningen Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Rita Nasti
- Stratingh Institute for Chemistry, University of Groningen Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Mael Poizat
- Symeres Kadijk 3 9747 AT Groningen The Netherlands
| | - Eric Buhler
- Laboratoire Matière et Systèmes Complexes (MSC), UMR CNRS 7057, Université Paris Cité Bâtiment Condorcet 75205 Paris Cedex 13 France
| | - Norbert Reiling
- RG Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center Borstel Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems Borstel Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1 66123 Saarbrücken Germany
- Helmholtz International Lab for Anti-infectives Campus Building E8.1 66123 Saarbrücken Germany
| | - Markus Fischer
- Hamburg School of Food Science, Institute of Food Chemistry Grindelallee 117 20146 Hamburg Germany
| | - Jean-Louis Reymond
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern Freiestrasse 3 3012 Bern Switzerland
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1 66123 Saarbrücken Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1 66123 Saarbrücken Germany
- Stratingh Institute for Chemistry, University of Groningen Nijenborgh 7 9747 AG Groningen The Netherlands
- Helmholtz International Lab for Anti-infectives Campus Building E8.1 66123 Saarbrücken Germany
| |
Collapse
|
19
|
Pernaute-Lau L, Camara M, Nóbrega de Sousa T, Morris U, Ferreira MU, Gil JP. An update on pharmacogenetic factors influencing the metabolism and toxicity of artemisinin-based combination therapy in the treatment of malaria. Expert Opin Drug Metab Toxicol 2022; 18:39-59. [PMID: 35285373 DOI: 10.1080/17425255.2022.2049235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Artemisinin-based combination therapies (ACTs) are recommended first-line antimalarials for uncomplicated Plasmodium falciparum malaria. Pharmacokinetic/pharmacodynamic variation associated with ACT drugs and their effect is documented. It is accepted to an extent that inter-individual variation is genetically driven, and should be explored for optimized antimalarial use. AREAS COVERED We provide an update on the pharmacogenetics of ACT antimalarial disposition. Beyond presently used antimalarials, we also refer to information available for the most notable next-generation drugs under development. The bibliographic approach was based on multiple Boolean searches on PubMed covering all recent publications since our previous review. EXPERT OPINION The last 10 years have witnessed an increase in our knowledge of ACT pharmacogenetics, including the first clear examples of its contribution as an exacerbating factor for drug-drug interactions. This knowledge gap is still large and is likely to widen as a new wave of antimalarial drug is looming, with few studies addressing their pharmacogenetics. Clinically useful pharmacogenetic markers are still not available, in particular, from an individual precision medicine perspective. A better understanding of the genetic makeup of target populations can be valuable for aiding decisions on mass drug administration implementation concerning region-specific antimalarial drug and dosage options.
Collapse
Affiliation(s)
- Leyre Pernaute-Lau
- Department of Microbiology, Tumor and Cell biology, Karolinska Institutet, Solna, Sweden.,Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisbon, Lisbon, 1749-016, Portugal
| | - Mahamadou Camara
- Department of Epidemiology of Parasitic Diseases, Faculty of Pharmacy, Malaria Research and Training Center, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Taís Nóbrega de Sousa
- Molecular Biology and Malaria Immunology Research Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brasil
| | - Ulrika Morris
- Department of Microbiology, Tumor and Cell biology, Karolinska Institutet, Solna, Sweden
| | - Marcelo Urbano Ferreira
- Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisbon, Lisbon, 1749-016, Portugal.,Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José Pedro Gil
- Department of Microbiology, Tumor and Cell biology, Karolinska Institutet, Solna, Sweden.,Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisbon, Lisbon, 1749-016, Portugal.,Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, Nova University of Lisbon, Portugal
| |
Collapse
|
20
|
Endo T, Takemae H, Sharma I, Furuya T. Multipurpose Drugs Active Against Both Plasmodium spp. and Microorganisms: Potential Application for New Drug Development. Front Cell Infect Microbiol 2021; 11:797509. [PMID: 35004357 PMCID: PMC8740689 DOI: 10.3389/fcimb.2021.797509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/01/2021] [Indexed: 12/29/2022] Open
Abstract
Malaria, a disease caused by the protozoan parasites Plasmodium spp., is still causing serious problems in endemic regions in the world. Although the WHO recommends artemisinin combination therapies for the treatment of malaria patients, the emergence of artemisinin-resistant parasites has become a serious issue and underscores the need for the development of new antimalarial drugs. On the other hand, new and re-emergences of infectious diseases, such as the influenza pandemic, Ebola virus disease, and COVID-19, are urging the world to develop effective chemotherapeutic agents against the causative viruses, which are not achieved to the desired level yet. In this review article, we describe existing drugs which are active against both Plasmodium spp. and microorganisms including viruses, bacteria, and fungi. We also focus on the current knowledge about the mechanism of actions of these drugs. Our major aims of this article are to describe examples of drugs that kill both Plasmodium parasites and other microbes and to provide valuable information to help find new ideas for developing novel drugs, rather than merely augmenting already existing drug repurposing efforts.
Collapse
Affiliation(s)
- Takuro Endo
- Laboratory of Veterinary Infectious Diseases, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hitoshi Takemae
- Center for Infectious Disease Epidemiology and Prevention Research, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Indu Sharma
- Department of Biological Sciences, Hampton University, Hampton, VA, United States
| | - Tetsuya Furuya
- Laboratory of Veterinary Infectious Diseases, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
21
|
Nordmann T, Borrmann S, Ramharter M. Drug-induced hypersensitivity to artemisinin-based therapies for malaria. Trends Parasitol 2021; 38:136-146. [PMID: 34561157 DOI: 10.1016/j.pt.2021.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
In the early 2000s, artemisinin-based combination therapy (ACT) was introduced as first-line treatment for uncomplicated Plasmodium falciparum malaria in virtually all endemic countries. However, despite the well-known excellent tolerability of ACTs, hypersensitivity to artemisinin derivatives remains a repeatedly documented adverse drug reaction of still unknown frequency. The clinical features of an artemisinin-induced hypersensitivity reaction range from mild to life-threatening severity, and a significant number of cases may pass unnoticed. In this review, we discuss the medical importance of hypersensitivity to artemisinin derivatives and we review data on the presumed frequency and its potential underlying mechanisms. Furthermore, we advocate to make alternative non-artemisinin-based drugs available for patients who do not tolerate artemisinin derivatives and to continue investing in the development of novel non-artemisinin-based combination regimens.
Collapse
Affiliation(s)
- Tamara Nordmann
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Infection Research (DZIF), partner site Hamburg-Luebeck-Borstel, Hamburg, Germany
| | - Steffen Borrmann
- Institute for Tropical Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany; Centre de Recherches Médicale de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Michael Ramharter
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Infection Research (DZIF), partner site Hamburg-Luebeck-Borstel, Hamburg, Germany; Centre de Recherches Médicale de Lambaréné (CERMEL), Lambaréné, Gabon.
| |
Collapse
|
22
|
Efforts Made to Eliminate Drug-Resistant Malaria and Its Challenges. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5539544. [PMID: 34497848 PMCID: PMC8421183 DOI: 10.1155/2021/5539544] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023]
Abstract
Since 2000, a good deal of progress has been made in malaria control. However, there is still an unacceptably high burden of the disease and numerous challenges limiting advancement towards its elimination and ultimate eradication. Among the challenges is the antimalarial drug resistance, which has been documented for almost all antimalarial drugs in current use. As a result, the malaria research community is working on the modification of existing treatments as well as the discovery and development of new drugs to counter the resistance challenges. To this effect, many products are in the pipeline and expected to be marketed soon. In addition to drug and vaccine development, mass drug administration (MDA) is under scientific scrutiny as an important strategy for effective utilization of the developed products. This review discusses the challenges related to malaria elimination, ongoing approaches to tackle the impact of drug-resistant malaria, and upcoming antimalarial drugs.
Collapse
|
23
|
Memvanga PB, Nkanga CI. Liposomes for malaria management: the evolution from 1980 to 2020. Malar J 2021; 20:327. [PMID: 34315484 PMCID: PMC8313885 DOI: 10.1186/s12936-021-03858-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/16/2021] [Indexed: 12/31/2022] Open
Abstract
Malaria is one of the most prevalent parasitic diseases and the foremost cause of morbidity in the tropical regions of the world. Strategies for the efficient management of this parasitic infection include adequate treatment with anti-malarial therapeutics and vaccination. However, the emergence and spread of resistant strains of malaria parasites to the majority of presently used anti-malarial medications, on the other hand, complicates malaria treatment. Other shortcomings of anti-malarial drugs include poor aqueous solubility, low permeability, poor bioavailability, and non-specific targeting of intracellular parasites, resulting in high dose requirements and toxic side effects. To address these limitations, liposome-based nanotechnology has been extensively explored as a new solution in malaria management. Liposome technology improves anti-malarial drug encapsulation, bioavailability, target delivery, and controlled release, resulting in increased effectiveness, reduced resistance progression, and fewer adverse effects. Furthermore, liposomes are exploited as immunological adjuvants and antigen carriers to boost the preventive effectiveness of malaria vaccine candidates. The present review discusses the findings from studies conducted over the last 40 years (1980-2020) using in vitro and in vivo settings to assess the prophylactic and curative anti-malarial potential of liposomes containing anti-malarial agents or antigens. This paper and the discussion herein provide a useful resource for further complementary investigations and may pave the way for the research and development of several available and affordable anti-malarial-based liposomes and liposomal malaria vaccines by allowing a thorough evaluation of liposomes developed to date for the management of malaria.
Collapse
Affiliation(s)
- Patrick B Memvanga
- Faculty of Pharmaceutical Sciences, Laboratory of Pharmaceutics and Phytopharmaceutical Drug Development, University of Kinshasa, B.P. 212, Kinshasa XI, Democratic Republic of the Congo.
| | - Christian I Nkanga
- Faculty of Pharmaceutical Sciences, Laboratory of Pharmaceutics and Phytopharmaceutical Drug Development, University of Kinshasa, B.P. 212, Kinshasa XI, Democratic Republic of the Congo
| |
Collapse
|
24
|
Saraiva RG, Dimopoulos G. Bacterial natural products in the fight against mosquito-transmitted tropical diseases. Nat Prod Rep 2021; 37:338-354. [PMID: 31544193 DOI: 10.1039/c9np00042a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Covering: up to 2019 Secondary metabolites of microbial origin have long been acknowledged as medically relevant, but their full potential remains largely unexploited. Of the countless natural compounds discovered thus far, only 5-10% have been isolated from microorganisms. At the same time, while whole-genome sequencing has demonstrated that bacteria and fungi often encode natural products, only a few genera have yet been mined for new compounds. This review explores the contributions of bacterial natural products to combatting infection by malaria parasites, filarial worms, and arboviruses such as dengue, Zika, Chikungunya, and West Nile. It highlights how molecules isolated from microorganisms ranging from marine cyanobacteria to mosquito endosymbionts can be exploited as antimicrobials and antivirals. Pursuit of this mostly untapped source of chemical entities will potentially result in new interventions against these tropical diseases, which are urgently needed to combat the increase in the incidence of resistance.
Collapse
Affiliation(s)
- Raúl G Saraiva
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA.
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
25
|
Shiomi K. Antiparasitic antibiotics from Japan. Parasitol Int 2021; 82:102298. [PMID: 33548522 DOI: 10.1016/j.parint.2021.102298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/19/2020] [Accepted: 01/27/2021] [Indexed: 11/29/2022]
Abstract
Antibiotics are microbial secondary metabolites and they are important for the treatment of infectious diseases. Japanese researchers have made a large contribution to studies of antibiotics, and they have also been important in the discovery of antiparasitic antibiotics. Satoshi Ōmura received the Nobel Prize in 2015 for the "discoveries concerning a novel therapy against infections caused by roundworm parasites", which means discovery of a new nematocidal antibiotic, avermectin. Here, I review the many antiparasitic antibiotics and their lead compounds that have been discovered for use in human and veterinary medicine.
Collapse
Affiliation(s)
- Kazuro Shiomi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan.
| |
Collapse
|
26
|
Development of sustainable research excellence with a global perspective on infectious diseases: Centre de Recherches Médicales de Lambaréné (CERMEL), Gabon. Wien Klin Wochenschr 2021; 133:500-508. [PMID: 33398458 PMCID: PMC7781170 DOI: 10.1007/s00508-020-01794-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/30/2020] [Indexed: 11/17/2022]
Abstract
Medical research in sub-Saharan Africa is of high priority for societies to respond adequately to local health needs. Often enough it remains a challenge to build up capacity in infrastructure and human resources to highest international standards and to sustain this over mid-term to long-term periods due to difficulties in obtaining long-term institutional core funding, attracting highly qualified scientists for medical research and coping with ever changing structural and political environments. The Centre de Recherches Médicales de Lambaréné (CERMEL) serves as model for how to overcome such challenges and to continuously increase its impact on medical care in Central Africa and beyond. Starting off as a research annex to the Albert Schweitzer Hospital in Lambaréné, Gabon, it has since then expanded its activities to academic and regulatory clinical trials for drugs, vaccines and diagnostics in the field of malaria, tuberculosis, and a wide range of poverty related and neglected tropical infectious diseases. Advancing bioethics in medical research in Africa and steadily improving its global networks and infrastructures, CERMEL serves as a reference centre for several international consortia. In close collaboration with national authorities, CERMEL has become one of the main training hubs for medical research in Central Africa. It is hoped that CERMEL and its leitmotiv “to improve medical care for local populations” will serve as an inspiration to other institutions in sub-Saharan Africa to further increase African capacity to advance medicine.
Collapse
|
27
|
Van Long B, Allen G, Brauny M, Linh LTK, Pallerla SR, Huyen TTT, Van Tong H, Toan NL, Quyet D, Son HA, Velavan TP. Molecular surveillance and temporal monitoring of malaria parasites in focal Vietnamese provinces. Malar J 2020; 19:458. [PMID: 33384023 PMCID: PMC7775626 DOI: 10.1186/s12936-020-03561-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While the World Health Organization (WHO) Southeast Asia region has the second highest incidence of malaria worldwide, malaria in Vietnam is focal to few provinces, where delayed parasite clearance to anti-malarial drugs is documented. This study aims to understand Plasmodium species distribution and the genetic diversity of msp1 and msp2 of parasite populations using molecular tools. METHODS A total of 222 clinical isolates from individuals with uncomplicated malaria were subjected to Plasmodium species identification by nested real-time PCR. 166 isolates positive for Plasmodium falciparum mono infections were further genotyped for msp1 (MAD20, K1, and RO33), and msp2 allelic families (3D7 and FC27). Amplicons were resolved through capillary electrophoresis in the QIAxcel Advanced system. RESULTS Mono-infections were high and with 75% P. falciparum, 14% Plasmodium vivax and 9% P. falciparum/P. vivax co-infections, with less than 1% Plasmodium malariae identified. For msp1, MAD20 was the most prevalent (99%), followed by K1 (46%) allelic family, with no sample testing positive for RO33 (0%). For msp2, 3D7 allelic family was predominant (97%), followed by FC27 (10%). The multiplicity of infection of msp1 and msp2 was 2.6 and 1.1, respectively, and the mean overall multiplicity of infection was 3.7, with the total number of alleles ranging from 1 to 7. CONCLUSIONS Given the increasing importance of antimalarial drugs in the region, the genetic diversity of P. falciparum msp1 and msp2 should be regularly monitored with respect to treatment outcomes and/or efficacy studies in regions, where there are ongoing changes in the malaria epidemiology.
Collapse
Affiliation(s)
- Bui Van Long
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Genevieve Allen
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Melanie Brauny
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Le Thi Kieu Linh
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany.,Vietnamese-German Centre for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Srinivas Reddy Pallerla
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Tran Thi Thu Huyen
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Hoang Van Tong
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam.,Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nguyen Linh Toan
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Do Quyet
- Vietnam Military Medical University, Hanoi, Vietnam
| | - Ho Anh Son
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam.
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany. .,Vietnamese-German Centre for Medical Research (VG-CARE), Hanoi, Vietnam.
| |
Collapse
|
28
|
Non-hydroxamate inhibitors of 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR): A critical review and future perspective. Eur J Med Chem 2020; 213:113055. [PMID: 33303239 DOI: 10.1016/j.ejmech.2020.113055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022]
Abstract
1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR) catalyzes the second step of the non-mevalonate (or MEP) pathway that functions in several organisms and plants for the synthesis of isoprenoids. DXR is essential for the survival of multiple pathogenic bacteria/parasites, including those that cause tuberculosis and malaria in humans. DXR function is inhibited by fosmidomycin (1), a natural product, which forms a chelate with the active site divalent metal (Mg2+/Mn2+) through its hydroxamate metal-binding group (MBG). Most of the potent DXR inhibitors are structurally similar to 1 and retain hydroxamate despite the unfavourable pharmacokinetic and toxicity profile of the latter. We provide our perspective on the lack of non-hydroxamate DXR inhibitors. We also highlight the fundamental flaws in the design of MBG in these molecules, primarily responsible for their failure to inhibit DXR. We also suggest that for designing next-generation non-hydroxamate DXR inhibitors, approaches followed for other metalloenzymes targets may be exploited.
Collapse
|
29
|
Palmer LD, Minor KE, Mettlach JA, Rivera ES, Boyd KL, Caprioli RM, Spraggins JM, Dalebroux ZD, Skaar EP. Modulating Isoprenoid Biosynthesis Increases Lipooligosaccharides and Restores Acinetobacter baumannii Resistance to Host and Antibiotic Stress. Cell Rep 2020; 32:108129. [PMID: 32905776 PMCID: PMC7519801 DOI: 10.1016/j.celrep.2020.108129] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/19/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Acinetobacter baumannii is a leading cause of ventilator-associated pneumonia and a critical threat due to multidrug resistance. The A. baumannii outer membrane is an asymmetric lipid bilayer composed of inner leaflet glycerophospholipids and outer leaflet lipooligosaccharides. Deleting mlaF of the maintenance of lipid asymmetry (Mla) system causes A. baumannii to become more susceptible to pulmonary surfactants and antibiotics and decreases bacterial survival in the lungs of mice. Spontaneous suppressor mutants isolated from infected mice contain an ISAba11 insertion upstream of the ispB initiation codon, an essential isoprenoid biosynthesis gene. The insertion restores antimicrobial resistance and virulence to ΔmlaF. The suppressor strain increases lipooligosaccharides, suggesting that the mechanism involves balancing the glycerophospholipids/lipooligosaccharides ratio on the bacterial surface. An identical insertion exists in an extensively drug-resistant A. baumannii isolate, demonstrating its clinical relevance. These data show that the stresses bacteria encounter during infection select for genomic rearrangements that increase resistance to antimicrobials.
Collapse
Affiliation(s)
- Lauren D Palmer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Keaton E Minor
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Joshua A Mettlach
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Emilio S Rivera
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Richard M Caprioli
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey M Spraggins
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Zachary D Dalebroux
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
30
|
Abstract
The scientific community worldwide has realized that malaria elimination will not be possible without development of safe and effective transmission-blocking interventions. Primaquine, the only WHO recommended transmission-blocking drug, is not extensively utilized because of the toxicity issues in G6PD deficient individuals. Therefore, there is an urgent need to develop novel therapeutic interventions that can target malaria parasites and effectively block transmission. But at first, it is imperative to unravel the existing portfolio of transmission-blocking drugs. This review highlights transmission-blocking potential of current antimalarial drugs and drugs that are in various stages of clinical development. The collective analysis of the relationships between the structure and the activity of transmission-blocking drugs is expected to help in the design of new transmission-blocking antimalarials.
Collapse
|
31
|
Zoleko Manego R, Koehne E, Kreidenweiss A, Nzigou Mombo B, Adegbite BR, Dimessa Mbadinga LB, Akinosho M, Matthewman J, Adegnika AA, Ramharter M, Mombo-Ngoma G. Description of Plasmodium falciparum infections in central Gabon demonstrating high parasite densities among symptomatic adolescents and adults. Malar J 2019; 18:371. [PMID: 31752891 PMCID: PMC6873720 DOI: 10.1186/s12936-019-3002-9] [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: 07/10/2019] [Accepted: 11/11/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Malaria remains a public health issue, particularly in sub-Saharan Africa with special features of seriousness in young children and pregnant women. Adolescents and adults are reported to have acquired a semi-immune status and, therefore, present with low parasitaemia. Children are understood to present with a much higher parasitaemia and severe malaria. It is a concern that effective malaria control programmes targeting young children may lead to a delay in the acquisition of acquired immunity and, therefore, causing a shift in the epidemiology of malaria. Prevalence and parasitaemia were explored in adolescents and adults with Plasmodium falciparum infections compared to young children in the area of Lambaréné, Gabon as an indicator for semi-immunity. METHODS A cross-sectional study was conducted at the Centre de Recherches Médicales de Lambaréné (CERMEL) during a 6-month period in 2018. Symptomatic patients, of all ages were screened for malaria at health facilities in Lambaréné and Fougamou and their respective surrounding villages in the central region of Gabon. Plasmodium falciparum infections were determined either by rapid diagnostic test (RDT) or by microscopy. Descriptive analysis of data on parasite densities, anaemia, and fever are presented. RESULTS 1589 individuals screened were included in this analysis, including 731 (46%) adolescents and adults. Out of 1377 assessed, the proportion of P. falciparum positive RDTs was high among adolescents (68%) and adults (44%), compared to young children (55%) and school children (72%). Out of 274 participants assessed for malaria by microscopy, 45 (16%) had a parasite count above 10,000/µl of which 9 (20%) were adults. CONCLUSION This study shows a high rate of P. falciparum infections in adolescents and adults associated with high-level parasitaemia similar to that of young children. Adolescents and adults seem to be an at-risk population, suggesting that malaria programmes should consider adolescents and adults during the implementation of malaria prevention and case management programmes with continuous care, since they also act as reservoirs for P. falciparum.
Collapse
Affiliation(s)
- Rella Zoleko Manego
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Wilhelmstrasse 27, 72074, Tübingen, Germany.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I Dep. of Medicine, University Medical Center Hamburg-Eppendorf, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany
| | - Erik Koehne
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Andrea Kreidenweiss
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | | | | | | | - Malik Akinosho
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon
| | - Julian Matthewman
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon
| | - Ayola Akim Adegnika
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Michael Ramharter
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I Dep. of Medicine, University Medical Center Hamburg-Eppendorf, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany
| | - Ghyslain Mombo-Ngoma
- Centre de Recherches Médicales de Lambaréné, B.P. 242, Lambaréné, Gabon. .,Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Wilhelmstrasse 27, 72074, Tübingen, Germany. .,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I Dep. of Medicine, University Medical Center Hamburg-Eppendorf, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany.
| |
Collapse
|
32
|
Lienau C, Gräwert T, Alves Avelar LA, Illarionov B, Held J, Knaab TC, Lungerich B, van Geelen L, Meier D, Geissler S, Cynis H, Riederer U, Buchholz M, Kalscheuer R, Bacher A, Mordmüller B, Fischer M, Kurz T. Novel reverse thia-analogs of fosmidomycin: Synthesis and antiplasmodial activity. Eur J Med Chem 2019; 181:111555. [DOI: 10.1016/j.ejmech.2019.07.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 01/17/2023]
|
33
|
Fosmidomycin, an inhibitor of isoprenoid synthesis, induces persistence in Chlamydia by inhibiting peptidoglycan assembly. PLoS Pathog 2019; 15:e1008078. [PMID: 31622442 PMCID: PMC6818789 DOI: 10.1371/journal.ppat.1008078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/29/2019] [Accepted: 09/12/2019] [Indexed: 11/19/2022] Open
Abstract
The antibiotic, fosmidomycin (FSM) targets the methylerythritol phosphate (MEP) pathway of isoprenoid synthesis by inhibiting the essential enzyme, 1-deoxy-D-xylulose 5-phosphate reductoisomerase (Dxr) and is lethal to intracellular parasites and bacteria. The obligate intracellular bacterial pathogen, Chlamydia trachomatis, alternates between two developmental forms: the extracellular, infectious elementary body (EB), and the intracellular, replicative form called the reticulate body (RB). Several stressful growth conditions including iron deprivation halt chlamydial cell division and cause development of a morphologically enlarged, but viable form termed an aberrant body (AB). This phenotype constitutes the chlamydial developmental state known as persistence. This state is reversible as removal of the stressor allows the chlamydiae to re-enter and complete the normal developmental cycle. Bioinformatic analysis indicates that C. trachomatis encodes a homolog of Dxr, but its function and the requirement for isoprenoid synthesis in chlamydial development is not fully understood. We hypothesized that chlamydial Dxr (DxrCT) is functional and that the methylerythritol phosphate (MEP) pathway is required for normal chlamydial development. Thus, FSM exposure should be lethal to C. trachomatis. Overexpression of chlamydial Dxr (DxrCT) in Escherichia coli under FSM exposure and in a conditionally lethal dxr mutant demonstrated that DxrCT functions similarly to E. coli Dxr. When Chlamydia-infected cultures were exposed to FSM, EB production was significantly reduced. However, titer recovery assays, electron microscopy, and peptidoglycan labeling revealed that FSM inhibition of isoprenoid synthesis is not lethal to C. trachomatis, but instead induces persistence. Bactoprenol is a critical isoprenoid required for peptidoglycan precursor assembly. We therefore conclude that FSM induces persistence in Chlamydia by preventing bactoprenol production necessary for peptidoglycan precursor assembly and subsequent cell division.
Collapse
|
34
|
Mengue JB, Held J, Kreidenweiss A. AQ-13 - an investigational antimalarial drug. Expert Opin Investig Drugs 2019; 28:217-222. [DOI: 10.1080/13543784.2019.1560419] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Juliana Boex Mengue
- Institute of Tropical Medicine, University Hospital Tuebingen, Tuebingen,
Germany
| | - Jana Held
- Institute of Tropical Medicine, University Hospital Tuebingen, Tuebingen,
Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University Hospital Tuebingen, Tuebingen,
Germany
- Centre de Recherches Médicales de Lambaréné, Albert Schweitzer Hospital,
Lambaréné, Gabon
| |
Collapse
|
35
|
Courtens C, Risseeuw M, Caljon G, Cos P, Van Calenbergh S. Acyloxybenzyl and Alkoxyalkyl Prodrugs of a Fosmidomycin Surrogate as Antimalarial and Antitubercular Agents. ACS Med Chem Lett 2018; 9:986-989. [PMID: 30344904 DOI: 10.1021/acsmedchemlett.8b00223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Two classes of prodrugs of a fosmidomycin surrogate were synthesized and investigated for their ability to inhibit in vitro growth of P. falciparum and M. tuberculosis. To this end, a novel efficient synthesis route was developed involving a cross metathesis reaction as a key step. Alkoxyalkyl prodrugs show decent antimalarial activities, but acyloxybenzyl prodrugs proved to be the most interesting and show enhanced antimalarial and antitubercular activity. The most active antimalarial analogues show low nanomolar IC50 values.
Collapse
Affiliation(s)
- Charlotte Courtens
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Martijn Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Guy Caljon
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| |
Collapse
|
36
|
Abstract
The last two decades have seen a surge in antimalarial drug development with product development partnerships taking a leading role. Resistance of Plasmodium falciparum to the artemisinin derivatives, piperaquine and mefloquine in Southeast Asia means new antimalarials are needed with some urgency. There are at least 13 agents in clinical development. Most of these are blood schizonticides for the treatment of uncomplicated falciparum malaria, under evaluation either singly or as part of two-drug combinations. Leading candidates progressing through the pipeline are artefenomel-ferroquine and lumefantrine-KAF156, both in Phase 2b. Treatment of severe malaria continues to rely on two parenteral drugs with ancient forebears: artesunate and quinine, with sevuparin being evaluated as an adjuvant therapy. Tafenoquine is under review by stringent regulatory authorities for approval as a single-dose treatment for Plasmodium vivax relapse prevention. This represents an advance over standard 14-day primaquine regimens; however, the risk of acute haemolytic anaemia in patients with glucose-6-phosphate dehydrogenase deficiency remains. For disease prevention, several of the newer agents show potential but are unlikely to be recommended for use in the main target groups of pregnant women and young children for some years. Latest predictions are that the malaria burden will continue to be high in the coming decades. This fact, coupled with the repeated loss of antimalarials to resistance, indicates that new antimalarials will be needed for years to come. Failure of the artemisinin-based combinations in Southeast Asia has stimulated a reappraisal of current approaches to combination therapy for malaria with incorporation of three or more drugs in a single treatment under consideration.
Collapse
Affiliation(s)
- Elizabeth A Ashley
- Myanmar Oxford Clinical Research Unit, Yangon, Myanmar.
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | | |
Collapse
|