1
|
Bhambid M, Walunj SB, Anupama CA, Jain S, Mehta D, Arya A, Wagstaff KM, Panda A, Jans DA, Mohmmed A, Patankar S. Importin α inhibitors act against the differentiated stages of apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii. J Antimicrob Chemother 2025; 80:485-495. [PMID: 39691987 DOI: 10.1093/jac/dkae434] [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: 07/15/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Nuclear import, dependent on the transporter importin α (IMPα), is a drug target for apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii. Indeed, a panel of small molecule inhibit interactions between IMPα and nuclear localization signals (NLSs) in vitro and the growth of rapidly dividing stages (P. falciparum blood stages and T. gondii tachyzoites) in culture. OBJECTIVES As new drugs targeting multiple life cycle stages of both parasites are required, the panel of IMPα inhibitors was tested for their ability to inhibit nuclear transport in the rapidly dividing stages and the maturation of differentiated stages (P. falciparum gametocytes and T. gondii bradyzoites). METHODS Using biophysical assays, Bay 11-7082, a Bay 11-7085 structural analogue, was tested for inhibition of IMPα:NLS interactions. The effect of the panel of inhibitors on the nuclear localization of reporter proteins was analysed in both parasites using transfections and microscopy. Also, using microscopy, the effect of inhibitors on differentiated stages of both parasites was tested. RESULTS Bay 11-7085 can inhibit nuclear transport in tachyzoites, while GW5074 and Caffeic Acid Phenethyl Ester (CAPE) can inhibit nuclear transport in the blood stages. Interestingly, CAPE can strongly inhibit gametocyte maturation, and Bay 11-7082 and Bay 11-7085 weakly inhibit bradyzoite differentiation. CONCLUSIONS As differentiation of gametocytes and bradyzoites is dependent on the activation of gene expression triggered by the nuclear translocation of transcription factors, our work provides a 'proof of concept' that targeting nuclear import is a viable strategy for the development of therapeutics against multiple stages of apicomplexan parasites, some of which are recalcitrant to existing drugs.
Collapse
Affiliation(s)
- Manasi Bhambid
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sujata B Walunj
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- IITB-Monash Research Academy, IIT Bombay, Mumbai, India
| | - C A Anupama
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shilpi Jain
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Diksha Mehta
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- IITB-Monash Research Academy, IIT Bombay, Mumbai, India
| | - Anjali Arya
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Ashutosh Panda
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - David A Jans
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Asif Mohmmed
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Swati Patankar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
2
|
Bravo P, Bizzarri L, Steinbrunn D, Lohse J, Hirsch AKH, Mäser P, Rottmann M, Hahne H. Integral Solvent-Induced Protein Precipitation for Target-Engagement Studies in Plasmodium falciparum. ACS Infect Dis 2024; 10:4073-4086. [PMID: 39631773 DOI: 10.1021/acsinfecdis.4c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The limited understanding of the mechanism of action (MoA) of several antimalarials and the rise of drug resistance toward existing malaria therapies emphasizes the need for new strategies to uncover the molecular target of compounds in Plasmodium falciparum. Integral solvent-induced protein precipitation (iSPP) is a quantitative mass spectrometry-based (LC-MS/MS) proteomics technique. The iSPP leverages the change in solvent-induced denaturation of the drug-bound protein relative to its unbound state, allowing identification of the direct drug-protein target without the need to modify the drug. Here, we demonstrate proof-of-concept of iSPP in P. falciparum (Pf) lysate. At first, we profiled the solvent-induced denaturation behavior of the Pf proteome, generating denaturation curves and determining the melting concentration (CM) of 2712 proteins. We then assessed the extent of stabilization of three antimalarial target proteins in multiple organic solvent gradients, allowing for a rational selection of an optimal solvent gradient. Subsequently, we validated iSPP by successfully showing target-engagement of several standard antimalarials. The iSPP assay allows the testing of multiple conditions within reasonable LC-MS/MS measurement time. Furthermore, it requires a minimal amount of protein input, reducing culturing time and simplifying protein extraction. We envision that iSPP will be useful as a complementary tool for MoA studies for next-generation antimalarials.
Collapse
Affiliation(s)
- Patricia Bravo
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Lorenzo Bizzarri
- OmicScouts GmbH, Lise-Meitner-Straße 30, D-85354 Freising, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, D-66123 Saarbrücken, Germany
| | - Dominik Steinbrunn
- OmicScouts GmbH, Lise-Meitner-Straße 30, D-85354 Freising, Germany
- TUM School of Natural Sciences, Department of Bioscience, Technical University of Munich, Center for Functional Protein Assemblies (CPA), D-85748 Garching bei München, Germany
| | - Jonas Lohse
- OmicScouts GmbH, Lise-Meitner-Straße 30, D-85354 Freising, Germany
| | - Anna K H Hirsch
- Department of Pharmacy, Saarland University, Campus E8.1, D-66123 Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8.1, D-66123 Saarbrücken, Germany
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Hannes Hahne
- OmicScouts GmbH, Lise-Meitner-Straße 30, D-85354 Freising, Germany
| |
Collapse
|
3
|
Appetecchia F, Fabbrizi E, Fiorentino F, Consalvi S, Biava M, Poce G, Rotili D. Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development. Pharmaceuticals (Basel) 2024; 17:962. [PMID: 39065810 PMCID: PMC11279868 DOI: 10.3390/ph17070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| |
Collapse
|
4
|
Giannangelo C, Challis MP, Siddiqui G, Edgar R, Malcolm TR, Webb CT, Drinkwater N, Vinh N, Macraild C, Counihan N, Duffy S, Wittlin S, Devine SM, Avery VM, De Koning-Ward T, Scammells P, McGowan S, Creek DJ. Chemoproteomics validates selective targeting of Plasmodium M1 alanyl aminopeptidase as an antimalarial strategy. eLife 2024; 13:RP92990. [PMID: 38976500 PMCID: PMC11230628 DOI: 10.7554/elife.92990] [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] [Indexed: 07/10/2024] Open
Abstract
New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum (PfA-M1) and Plasmodium vivax (PvA-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets PfA-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on PfA-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of PfA-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.
Collapse
Affiliation(s)
- Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Matthew P Challis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Rebecca Edgar
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Tess R Malcolm
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Chaille T Webb
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Nyssa Drinkwater
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Natalie Vinh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Christopher Macraild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Natalie Counihan
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Sandra Duffy
- Discovery Biology, Centre for Cellular Phenomics, Griffith UniversityNathanAustralia
| | - Sergio Wittlin
- Swiss Tropical and Public Health InstituteAllschwilSwitzerland
- University of BaselBaselSwitzerland
| | - Shane M Devine
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, The University of MelbourneParkvilleAustralia
| | - Vicky M Avery
- Discovery Biology, Centre for Cellular Phenomics, Griffith UniversityNathanAustralia
- School of Environment and Science, Griffith UniversityNathanAustralia
| | - Tania De Koning-Ward
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Peter Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Sheena McGowan
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| |
Collapse
|
5
|
Edgar RCS, Malcolm TR, Siddiqui G, Giannangelo C, Counihan NA, Challis M, Duffy S, Chowdhury M, Marfurt J, Dans M, Wirjanata G, Noviyanti R, Daware K, Suraweera CD, Price RN, Wittlin S, Avery VM, Drinkwater N, Charman SA, Creek DJ, de Koning-Ward TF, Scammells PJ, McGowan S. On-target, dual aminopeptidase inhibition provides cross-species antimalarial activity. mBio 2024; 15:e0096624. [PMID: 38717141 PMCID: PMC11237774 DOI: 10.1128/mbio.00966-24] [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: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 06/13/2024] Open
Abstract
To combat the global burden of malaria, development of new drugs to replace or complement current therapies is urgently required. Here, we show that the compound MMV1557817 is a selective, nanomolar inhibitor of both Plasmodium falciparum and Plasmodium vivax aminopeptidases M1 and M17, leading to inhibition of end-stage hemoglobin digestion in asexual parasites. MMV1557817 can kill sexual-stage P. falciparum, is active against murine malaria, and does not show any shift in activity against a panel of parasites resistant to other antimalarials. MMV1557817-resistant P. falciparum exhibited a slow growth rate that was quickly outcompeted by wild-type parasites and were sensitized to the current clinical drug, artemisinin. Overall, these results confirm MMV1557817 as a lead compound for further drug development and highlights the potential of dual inhibition of M1 and M17 as an effective multi-species drug-targeting strategy.IMPORTANCEEach year, malaria infects approximately 240 million people and causes over 600,000 deaths, mostly in children under 5 years of age. For the past decade, artemisinin-based combination therapies have been recommended by the World Health Organization as the standard malaria treatment worldwide. Their widespread use has led to the development of artemisinin resistance in the form of delayed parasite clearance, alongside the rise of partner drug resistance. There is an urgent need to develop and deploy new antimalarial agents with novel targets and mechanisms of action. Here, we report a new and potent antimalarial compound, known as MMV1557817, and show that it targets multiple stages of the malaria parasite lifecycle, is active in a preliminary mouse malaria model, and has a novel mechanism of action. Excitingly, resistance to MMV15578117 appears to be self-limiting, suggesting that development of the compound may provide a new class of antimalarial.
Collapse
Affiliation(s)
- Rebecca C S Edgar
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Tess R Malcolm
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Natalie A Counihan
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Matthew Challis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Sandra Duffy
- Discovery Biology, Centre for Cellular Phenomics, Griffith University, Nathan, Queensland, Australia
| | - Mrittika Chowdhury
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Jutta Marfurt
- Global Health and Tropical Medicine Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Madeline Dans
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Grennady Wirjanata
- Global Health and Tropical Medicine Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | | | - Kajal Daware
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Chathura D Suraweera
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Ric N Price
- Global Health and Tropical Medicine Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Vicky M Avery
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, Queensland, Australia
| | - Nyssa Drinkwater
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Sheena McGowan
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
6
|
Creek D, Giannangelo C, Challis M, Siddiqui G, Edgar R, Malcolm T, Webb C, Drinkwater N, Vinh N, MacRaild C, Counihan N, Duffy S, Wittlin S, Devine S, Avery V, de Koning-Ward T, Scammells P, McGowan S. Chemoproteomics validates selective targeting of Plasmodium M1 alanyl aminopeptidase as an antimalarial strategy. RESEARCH SQUARE 2024:rs.3.rs-3251230. [PMID: 38746424 PMCID: PMC11092810 DOI: 10.21203/rs.3.rs-3251230/v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum ( Pf A-M1) and Plasmodium vivax ( Pv A-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets Pf A-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on Pf A-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of Pf A-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.
Collapse
|
7
|
Basco LK. Cultivation of Asexual Intraerythrocytic Stages of Plasmodium falciparum. Pathogens 2023; 12:900. [PMID: 37513747 PMCID: PMC10384318 DOI: 10.3390/pathogens12070900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Successfully developed in 1976, the continuous in vitro culture of Plasmodium falciparum has many applications in the field of malaria research. It has become an important experimental model that directly uses a human pathogen responsible for a high prevalence of morbidity and mortality in many parts of the world and is a major source of biological material for immunological, biochemical, molecular, and pharmacological studies. Until present, the basic techniques described by Trager and Jensen and Haynes et al. remain unchanged in many malaria research laboratories. Nonetheless, different factors, including culture media, buffers, serum substitutes and supplements, sources of erythrocytes, and conditions of incubation (especially oxygen concentration), have been modified by different investigators to adapt the original technique in their laboratories or enhance the in vitro growth of the parasites. The possible effects and benefits of these modifications for the continuous cultivation of asexual intraerythrocytic stages of P. falciparum, as well as future challenges in developing a serum-free cultivation system and axenic cultures, are discussed.
Collapse
Affiliation(s)
- Leonardo K Basco
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Service de Santé des Armées (SSA), Unité Mixte de Recherche (UMR) Vecteurs-Infections Tropicales et Méditerranéennes (VITROME), 13005 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
| |
Collapse
|
8
|
Nkhoma SC, Ahmed AOA, Porier D, Rashid S, Bradford R, Molestina RE, Stedman TT. Dynamics of parasite growth in genetically diverse Plasmodium falciparum isolates. Mol Biochem Parasitol 2023; 254:111552. [PMID: 36731750 PMCID: PMC10149587 DOI: 10.1016/j.molbiopara.2023.111552] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/24/2022] [Accepted: 01/26/2023] [Indexed: 02/01/2023]
Abstract
Multiple parasite lineages with different proliferation rates or fitness may coexist within a clinical malaria isolate, resulting in complex growth interactions and variations in phenotype. To elucidate the dynamics of parasite growth in multiclonal isolates, we measured growth rates (GRs) of three Plasmodium falciparum Cambodian isolates, including IPC_3445 (MRA-1236), IPC_5202 (MRA-1240), IPC_6403 (MRA-1285), and parasite lineages previously cloned from each of these isolates by limiting dilution. Following synchronization, in vitro cultures of each parasite line were maintained over four consecutive asexual cycles (192 h), with thin smears prepared at each 48-h cycle to estimate GR and fold change in parasitemia (FCP). Cell cycle time (CCT), the duration it takes for ring-stage parasites to develop into mature schizonts, was measured by monitoring the development of 0-3-h post-invasion rings for up to 52 h post-incubation. Laboratory lines 3D7 (MRA-102) and Dd2 (MRA-150) were used as controls. Significant differences in GR, FCP, and CCT were observed between parasite isolates and clonal lineages from each isolate. The parasite lines studied here have well-defined growth phenotypes and will facilitate basic malaria research and development of novel malaria interventions. These lines are available to malaria researchers through the MR4 collection of NIAID's BEI Resources Program.
Collapse
Affiliation(s)
- Standwell C Nkhoma
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA.
| | - Amel O A Ahmed
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA
| | - Danielle Porier
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA
| | - Sujatha Rashid
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA
| | - Rebecca Bradford
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA
| | - Robert E Molestina
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA
| | - Timothy T Stedman
- BEI Resources, American Type Culture Collection, 10801 University Boulevard, Manassas, VA 20110-2209, USA
| |
Collapse
|
9
|
Carucci M, Duez J, Tarning J, García-Barbazán I, Fricot-Monsinjon A, Sissoko A, Dumas L, Gamallo P, Beher B, Amireault P, Dussiot M, Dao M, Hull MV, McNamara CW, Roussel C, Ndour PA, Sanz LM, Gamo FJ, Buffet P. Safe drugs with high potential to block malaria transmission revealed by a spleen-mimetic screening. Nat Commun 2023; 14:1951. [PMID: 37029122 PMCID: PMC10082216 DOI: 10.1038/s41467-023-37359-2] [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: 07/20/2022] [Accepted: 03/15/2023] [Indexed: 04/09/2023] Open
Abstract
Malaria parasites like Plasmodium falciparum multiply in red blood cells (RBC), which are cleared from the bloodstream by the spleen when their deformability is altered. Drug-induced stiffening of Plasmodium falciparum-infected RBC should therefore induce their elimination from the bloodstream. Here, based on this original mechanical approach, we identify safe drugs with strong potential to block the malaria transmission. By screening 13 555 compounds with spleen-mimetic microfilters, we identified 82 that target circulating transmissible form of P. falciparum. NITD609, an orally administered PfATPase inhibitor with known effects on P. falciparum, killed and stiffened transmission stages in vitro at nanomolar concentrations. Short exposures to TD-6450, an orally-administered NS5A hepatitis C virus inhibitor, stiffened transmission parasite stages and killed asexual stages in vitro at high nanomolar concentrations. A Phase 1 study in humans with a primary safety outcome and a secondary pharmacokinetics outcome ( https://clinicaltrials.gov , ID: NCT02022306) showed no severe adverse events either with single or multiple doses. Pharmacokinetic modelling showed that these concentrations can be reached in the plasma of subjects receiving short courses of TD-6450. This physiologically relevant screen identified multiple mechanisms of action, and safe drugs with strong potential as malaria transmission-blocking agents which could be rapidly tested in clinical trials.
Collapse
Affiliation(s)
- Mario Carucci
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | | | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 10400, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Irene García-Barbazán
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28222, Madrid, Spain
| | - Aurélie Fricot-Monsinjon
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Abdoulaye Sissoko
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Lucie Dumas
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Pablo Gamallo
- Global Health Medicines R&D, GlaxoSmith Kline (GSK), 28760, Tres Cantos, Spain
| | - Babette Beher
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Pascal Amireault
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
- Laboratory of cellular and molecular mechanisms of hematological disorders and therapeutic implications, INSERM, 75014, Paris, France
| | - Michael Dussiot
- Laboratory of cellular and molecular mechanisms of hematological disorders and therapeutic implications, INSERM, 75014, Paris, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, MA, 02139, Cambridge, USA
| | - Mitchell V Hull
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Camille Roussel
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Laboratoire d'Hématologie générale, Hôpital Universitaire Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015, Paris, France
| | - Papa Alioune Ndour
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Laura Maria Sanz
- Global Health Medicines R&D, GlaxoSmith Kline (GSK), 28760, Tres Cantos, Spain
| | | | - Pierre Buffet
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France.
- Department of Infectious & Tropical Disease, AP-HP, Necker Hospital, 75015, Paris, France.
- Centre Médical de l'Institut Pasteur (CMIP), Institut Pasteur, 75015, Paris, France.
| |
Collapse
|
10
|
Nardella F, Dobrescu I, Hassan H, Rodrigues F, Thiberge S, Mancio-Silva L, Tafit A, Jallet C, Cadet-Daniel V, Goussin S, Lorthiois A, Menon Y, Molinier N, Pechalrieu D, Long C, Sautel F, Matondo M, Duchateau M, Médard G, Witkowski B, Scherf A, Halby L, Arimondo PB. Hemisynthetic alkaloids derived from trilobine are antimalarials with sustained activity in multidrug-resistant Plasmodium falciparum. iScience 2023; 26:105940. [PMID: 36718363 PMCID: PMC9883252 DOI: 10.1016/j.isci.2023.105940] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/23/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Malaria eradication requires the development of new drugs to combat drug-resistant parasites. We identified bisbenzylisoquinoline alkaloids isolated from Cocculus hirsutus that are active against Plasmodium falciparum blood stages. Synthesis of a library of 94 hemi-synthetic derivatives allowed to identify compound 84 that kills multi-drug resistant clinical isolates in the nanomolar range (median IC50 ranging from 35 to 88 nM). Chemical optimization led to compound 125 with significantly improved preclinical properties. 125 delays the onset of parasitemia in Plasmodium berghei infected mice and inhibits P. falciparum transmission stages in vitro (culture assays), and in vivo using membrane feeding assay in the Anopheles stephensi vector. Compound 125 also impairs P. falciparum development in sporozoite-infected hepatocytes, in the low micromolar range. Finally, by chemical pull-down strategy, we characterized the parasite interactome with trilobine derivatives, identifying protein partners belonging to metabolic pathways that are not targeted by the actual antimalarial drugs or implicated in drug-resistance mechanisms.
Collapse
Affiliation(s)
- Flore Nardella
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Irina Dobrescu
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Haitham Hassan
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Fabien Rodrigues
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Sabine Thiberge
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France,Center for Production and Infection of Anopheles (CEPIA), Center for Animal Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Liliana Mancio-Silva
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Ambre Tafit
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Corinne Jallet
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Véronique Cadet-Daniel
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Stéphane Goussin
- Center for Production and Infection of Anopheles (CEPIA), Center for Animal Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Audrey Lorthiois
- Center for Production and Infection of Anopheles (CEPIA), Center for Animal Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Yoann Menon
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Nicolas Molinier
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Dany Pechalrieu
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Christophe Long
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - François Sautel
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, Université de Paris-Cité, CNRS USR 2000, 28 rue du Dr Roux, 75015 Paris, France
| | - Magalie Duchateau
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, Université de Paris-Cité, CNRS USR 2000, 28 rue du Dr Roux, 75015 Paris, France
| | - Guillaume Médard
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, 85354 Freising, Germany
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Artur Scherf
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France,Corresponding author
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Paola B. Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France,USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France,Corresponding author
| |
Collapse
|
11
|
Omorou R, Bin Sa'id I, Delves M, Severini C, Kouakou YI, Bienvenu AL, Picot S. Protocols for Plasmodium gametocyte production in vitro: an integrative review and analysis. Parasit Vectors 2022; 15:451. [PMID: 36471426 PMCID: PMC9720971 DOI: 10.1186/s13071-022-05566-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/02/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The production of Plasmodium gametocytes in vitro is a real challenge. Many protocols have been described, but few have resulted in the production of viable and infectious gametocytes in sufficient quantities to conduct research on-but not limited to-transmission-blocking drug and vaccine development. The aim of this review was to identify and discuss gametocyte production protocols that have been developed over the last two decades. METHODS We analyzed the original gametocyte production protocols published from 2000 onwards based on a literature search and a thorough review. A systematic review was performed of relevant articles identified in the PubMed, Web of Sciences and ScienceDirect databases. RESULTS A total 23 studies on the production of Plasmodium gametocytes were identified, 19 involving in vitro Plasmodium falciparum, one involving Plasmodium knowlesi and three involving ex vivo Plasmodium vivax. Of the in vitro studies, 90% used environmental stressors to trigger gametocytogenesis. Mature gametocytemia of up to 4% was reported. CONCLUSIONS Several biological parameters contribute to an optimal production in vitro of viable and infectious mature gametocytes. The knowledge gained from this systematic review on the molecular mechanisms involved in gametocytogenesis enables reproducible gametocyte protocols with transgenic parasite lines to be set up. This review highlights the need for additional gametocyte production protocols for Plasmodium species other than P. falciparum.
Collapse
Affiliation(s)
- Roukayatou Omorou
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE-University Lyon1, University of Lyon, 69100, Villeurbanne, France.
| | - Ibrahim Bin Sa'id
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE-University Lyon1, University of Lyon, 69100, Villeurbanne, France.,Institut Agama Islam Negeri (IAIN) Kediri, 64127, Kota Kediri, Jawa Timur, Indonesia
| | - Michael Delves
- Department of Infection Biology, Faculty of Infectious Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1A 7HT, UK
| | - Carlo Severini
- Dipartimento Di Malattie Infettive, Istituto Superiore Di Sanità, Rome, Italy
| | - Yobouet Ines Kouakou
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE-University Lyon1, University of Lyon, 69100, Villeurbanne, France
| | - Anne-Lise Bienvenu
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE-University Lyon1, University of Lyon, 69100, Villeurbanne, France.,Service Pharmacie, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Stephane Picot
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE-University Lyon1, University of Lyon, 69100, Villeurbanne, France.,Institut de Parasitologie Et Mycologie Médicale, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
12
|
Duffy S, Avery VM. Naturally Acquired Kelch13 Mutations in Plasmodium falciparum Strains Modulate In Vitro Ring-Stage Artemisinin-Based Drug Tolerance and Parasite Survival in Response to Hyperoxia. Microbiol Spectr 2022; 10:e0128221. [PMID: 36094220 PMCID: PMC9602862 DOI: 10.1128/spectrum.01282-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/25/2022] [Indexed: 12/30/2022] Open
Abstract
The ring-stage survival assay was utilized to assess the impact of physiological hyperoxic stress on dihydroartemisinin (DHA) tolerance for a panel of Plasmodium falciparum strains with and without Kelch13 mutations. Strains without naturally acquired Kelch13 mutations or the postulated genetic background associated with delayed parasite clearance time demonstrated reduced proliferation under hyperoxic conditions in the subsequent proliferation cycle. Dihydroartemisinin tolerance in three isolates with naturally acquired Kelch13 mutations but not two genetically manipulated laboratory strains was modulated by in vitro hyperoxic stress exposure of early-ring-stage parasites in the cycle before drug exposure. Reduced parasite tolerance to additional derivatives, including artemisinin, artesunate, and OZ277, was observed within the second proliferation cycle. OZ439 and epoxomicin completely prevented parasite survival under both hyperoxia and normoxic in vitro culture conditions, highlighting the unique relationship between DHA tolerance and Kelch13 mutation-associated genetic background. IMPORTANCE Artemisinin-based combination therapy (ACT) for treating malaria is under intense scrutiny following treatment failures in the Greater Mekong subregion of Asia. This is further compounded by the potential for extensive loss of life if treatment failures extend to the African continent. Although Plasmodium falciparum has become resistant to all antimalarial drugs, artemisinin "resistance" does not present in the same way as resistance to other antimalarial drugs. Instead, a partial resistance or tolerance is demonstrated, associated with the parasite's genetic profile and linked to a molecular marker referred to as K13. It is suggested that parasites may have adapted to drug treatment, as well as the presence of underlying population health issues such as hemoglobinopathies, and/or environmental pressures, resulting in parasite tolerance to ACT. Understanding parasite evolution and control of artemisinin tolerance will provide innovative approaches to mitigate the development of artemisinin tolerance and thereby artemisinin-based drug treatment failure and loss of life globally to malaria infections.
Collapse
Affiliation(s)
- Sandra Duffy
- Discovery Biology, Griffith University, Nathan, Queensland, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith University, Nathan, Queensland, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
13
|
Hanifian H, Nateghpour M, Motevalli Haghi A, Teimouri A, Razavi S, Fariver L. Development and optimizing a simple and cost-effective medium for in vitro culture of Plasmodium berghei-ANKA strain with conserving its infectivity in BALB/c mice. BMC Res Notes 2022; 15:56. [PMID: 35168649 PMCID: PMC8845400 DOI: 10.1186/s13104-022-05946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES The current culture system for P. berghei still requires modifications in consistency and long-term maintenance of parasites considering their pathogenicity in culture media. Therefore, this study designed to further improvement of culture conditions and designing a cost-effective culture medium with minimum changes in pathogenicity for in vitro culture of P. berghei. RESULTS Results indicated that the rate of parasitaemia in our modified method remained statistically stable between days one to seven (P = 0.07). The current modified cultivation method was more efficient in maintaining of parasites for further days. Furthermore, in current method the stability of parasitaemia rate during day1 to day7 was in better rate compared to that in Ronan Jambou et al. and the differences between two methods were statistically significant (P = 0.001). The virulence of cultivated parasites in our modified method remained similar to frozen stock parasites as positive control group. No significant differences were seen in survival time between two groups of mice those were infected with either cultivated parasites or stock freeze parasites (P = 0.39) with the mean survival time of 20.83 ± 3.84 and 19.66 ± 1.21 days, respectively. Herein, we achieved a simple, cost-effective and applicable technique for culture of P. berghei.
Collapse
Affiliation(s)
- Haleh Hanifian
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Nateghpour
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Hadith and Medicine, Research Centre of Quran, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afsaneh Motevalli Haghi
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Aref Teimouri
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepand Razavi
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Fariver
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Abstract
Malaria parasites need to cope with changing environmental conditions that require strong countermeasures to ensure pathogen survival in the human and mosquito hosts. The molecular mechanisms that protect Plasmodium falciparum homeostasis during the complex life cycle remain unknown. Here, we identify cytosine methylation of tRNAAsp (GTC) as being critical to maintain stable protein synthesis. Using conditional knockout (KO) of a member of the DNA methyltransferase family, called Pf-DNMT2, RNA bisulfite sequencing demonstrated the selective cytosine methylation of this enzyme of tRNAAsp (GTC) at position C38. Although no growth defect on parasite proliferation was observed, Pf-DNMT2KO parasites showed a selective downregulation of proteins with a GAC codon bias. This resulted in a significant shift in parasite metabolism, priming KO parasites for being more sensitive to various types of stress. Importantly, nutritional stress made tRNAAsp (GTC) sensitive to cleavage by an unknown nuclease and increased gametocyte production (>6-fold). Our study uncovers an epitranscriptomic mechanism that safeguards protein translation and homeostasis of sexual commitment in malaria parasites. IMPORTANCE P. falciparum is the most virulent malaria parasite species, accounting for the majority of the disease mortality and morbidity. Understanding how this pathogen is able to adapt to different cellular and environmental stressors during its complex life cycle is crucial in order to develop new strategies to tackle the disease. In this study, we identified the writer of a specific tRNA cytosine methylation site as a new layer of epitranscriptomic regulation in malaria parasites that regulates the translation of a subset of parasite proteins (>400) involved in different metabolic pathways. Our findings give insight into a novel molecular mechanism that regulates P. falciparum response to drug treatment and sexual commitment.
Collapse
|
15
|
Property activity refinement of 2-anilino 4-amino substituted quinazolines as antimalarials with fast acting asexual parasite activity. Bioorg Chem 2021; 117:105359. [PMID: 34689083 DOI: 10.1016/j.bioorg.2021.105359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/25/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022]
Abstract
Malaria is a devastating disease caused by Plasmodium parasites. Emerging resistance against current antimalarial therapeutics has engendered the need to develop antimalarials with novel structural classes. We recently described the identification and initial optimization of the 2-anilino quinazoline antimalarial class. Here, we refine the physicochemical properties of this antimalarial class with the aim to improve aqueous solubility and metabolism and to reduce adverse promiscuity. We show the physicochemical properties of this class are intricately balanced with asexual parasite activity and human cell cytotoxicity. Structural modifications we have implemented improved LipE, aqueous solubility and in vitro metabolism while preserving fast acting P. falciparum asexual stage activity. The lead compounds demonstrated equipotent activity against P. knowlesi parasites and were not predisposed to resistance mechanisms of clinically used antimalarials. The optimized compounds exhibited modest activity against early-stage gametocytes, but no activity against pre-erythrocytic liver parasites. Confoundingly, the refined physicochemical properties installed in the compounds did not engender improved oral efficacy in a P. berghei mouse model of malaria compared to earlier studies on the 2-anilino quinazoline class. This study provides the framework for further development of this antimalarial class.
Collapse
|
16
|
The Novel bis-1,2,4-Triazine MIPS-0004373 Demonstrates Rapid and Potent Activity against All Blood Stages of the Malaria Parasite. Antimicrob Agents Chemother 2021; 65:e0031121. [PMID: 34460304 DOI: 10.1128/aac.00311-21] [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] [Indexed: 11/20/2022] Open
Abstract
Novel bis-1,2,4-triazine compounds with potent in vitro activity against Plasmodium falciparum parasites were recently identified. The bis-1,2,4-triazines represent a unique antimalarial pharmacophore and are proposed to act by a novel but as-yet-unknown mechanism of action. This study investigated the activity of the bis-1,2,4-triazine MIPS-0004373 across the mammalian life cycle stages of the parasite and profiled the kinetics of activity against blood and transmission stage parasites in vitro and in vivo. MIPS-0004373 demonstrated rapid and potent activity against P. falciparum, with excellent in vitro activity against all asexual blood stages. Prolonged in vitro drug exposure failed to generate stable resistance de novo, suggesting a low propensity for the emergence of resistance. Excellent activity was observed against sexually committed ring stage parasites, but activity against mature gametocytes was limited to inhibiting male gametogenesis. Assessment of liver stage activity demonstrated good activity in an in vitro P. berghei model but no activity against Plasmodium cynomolgi hypnozoites or liver schizonts. The bis-1,2,4-triazine MIPS-0004373 efficiently cleared an established P. berghei infection in vivo, with efficacy similar to that of artesunate and chloroquine and a recrudescence profile comparable to that of chloroquine. This study demonstrates the suitability of bis-1,2,4-triazines for further development toward a novel treatment for acute malaria.
Collapse
|
17
|
Laleu B, Akao Y, Ochida A, Duffy S, Lucantoni L, Shackleford DM, Chen G, Katneni K, Chiu FCK, White KL, Chen X, Sturm A, Dechering KJ, Crespo B, Sanz LM, Wang B, Wittlin S, Charman SA, Avery VM, Cho N, Kamaura M. Discovery and Structure-Activity Relationships of Quinazolinone-2-carboxamide Derivatives as Novel Orally Efficacious Antimalarials. J Med Chem 2021; 64:12582-12602. [PMID: 34437804 DOI: 10.1021/acs.jmedchem.1c00441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A phenotypic high-throughput screen allowed discovery of quinazolinone-2-carboxamide derivatives as a novel antimalarial scaffold. Structure-activity relationship studies led to identification of a potent inhibitor 19f, 95-fold more potent than the original hit compound, active against laboratory-resistant strains of malaria. Profiling of 19f suggested a fast in vitro killing profile. In vivo activity in a murine model of human malaria in a dose-dependent manner constitutes a concomitant benefit.
Collapse
Affiliation(s)
- Benoît Laleu
- Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Yuichiro Akao
- Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Atsuko Ochida
- Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Sandra Duffy
- Discovery Biology, Griffith University, Brisbane Innovation Park, Don Young Road, Nathan 4111, Queensland, Australia
| | - Leonardo Lucantoni
- Discovery Biology, Griffith University, Brisbane Innovation Park, Don Young Road, Nathan 4111, Queensland, Australia
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Francis C K Chiu
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Karen L White
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Xue Chen
- WuXi AppTec (Wuhan) Company Ltd., 666 Gaoxin Avenue, Donghu New Technology Development Area, Wuhan 430075, China
| | - Angelika Sturm
- TropIQ Health Sciences, Transistorweg 5-C02, 6534 AT Nijmegen, The Netherlands
| | - Koen J Dechering
- TropIQ Health Sciences, Transistorweg 5-C02, 6534 AT Nijmegen, The Netherlands
| | - Benigno Crespo
- Global Health, GlaxoSmithKline R&D, Tres Cantos, 28760, Madrid, Spain
| | - Laura M Sanz
- Global Health, GlaxoSmithKline R&D, Tres Cantos, 28760, Madrid, Spain
| | - Binglin Wang
- WuXi AppTec (Wuhan) Company Ltd., 666 Gaoxin Avenue, Donghu New Technology Development Area, Wuhan 430075, China
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland.,University of Basel, 4002 Basel, Switzerland
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith University, Brisbane Innovation Park, Don Young Road, Nathan 4111, Queensland, Australia
| | - Nobuo Cho
- Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Kamaura
- Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
18
|
CRISPR/Cas9-engineered inducible gametocyte producer lines as a valuable tool for Plasmodium falciparum malaria transmission research. Nat Commun 2021; 12:4806. [PMID: 34376675 PMCID: PMC8355313 DOI: 10.1038/s41467-021-24954-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
The malaria parasite Plasmodium falciparum replicates inside erythrocytes in the blood of infected humans. During each replication cycle, a small proportion of parasites commits to sexual development and differentiates into gametocytes, which are essential for parasite transmission via the mosquito vector. Detailed molecular investigation of gametocyte biology and transmission has been hampered by difficulties in generating large numbers of these highly specialised cells. Here, we engineer P. falciparum NF54 inducible gametocyte producer (iGP) lines for the routine mass production of synchronous gametocytes via conditional overexpression of the sexual commitment factor GDV1. NF54/iGP lines consistently achieve sexual commitment rates of 75% and produce viable gametocytes that are transmissible by mosquitoes. We also demonstrate that further genetic engineering of NF54/iGP parasites is a valuable tool for the targeted exploration of gametocyte biology. In summary, we believe the iGP approach developed here will greatly expedite basic and applied malaria transmission stage research.
Collapse
|
19
|
Nardella F, Halby L, Dobrescu I, Viluma J, Bon C, Claes A, Cadet-Daniel V, Tafit A, Roesch C, Hammam E, Erdmann D, Mairet-Khedim M, Peronet R, Mecheri S, Witkowski B, Scherf A, Arimondo PB. Procainamide-SAHA Fused Inhibitors of hHDAC6 Tackle Multidrug-Resistant Malaria Parasites. J Med Chem 2021; 64:10403-10417. [PMID: 34185525 DOI: 10.1021/acs.jmedchem.1c00821] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetic post-translational modifications are essential for human malaria parasite survival and progression through its life cycle. Here, we present new functionalized suberoylanilide hydroxamic acid (SAHA) derivatives that chemically combine the pan-histone deacetylase inhibitor SAHA with the DNA methyltransferase inhibitor procainamide. A three- or four-step chemical synthesis was designed starting from cheap raw materials. Compared to the single drugs, the combined molecules showed a superior activity in Plasmodium and a potent inhibition against human HDAC6, exerting no cytotoxicity in human cell lines. These new compounds are fully active in multidrug-resistant Plasmodium falciparum Cambodian isolates. They target transmission of the parasite by inducing irreversible morphological changes in gametocytes and inhibiting exflagellation. The compounds are slow-acting and have an additive antimalarial effect in combination with fast-acting epidrugs and dihydroartemisinin. The lead compound decreases parasitemia in mice in a severe malaria model. Taken together, this novel fused molecule offers an affordable alternative to current failing antimalarial therapy.
Collapse
Affiliation(s)
- Flore Nardella
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Irina Dobrescu
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Johanna Viluma
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Corentin Bon
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France.,Ecole Doctorale MTCI ED563, Université de Paris, Sorbonne Paris Cité, Paris 75270, France
| | - Aurélie Claes
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Véronique Cadet-Daniel
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Ambre Tafit
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Camille Roesch
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Elie Hammam
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Diane Erdmann
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France.,Ecole Doctorale MTCI ED563, Université de Paris, Sorbonne Paris Cité, Paris 75270, France
| | - Melissa Mairet-Khedim
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Roger Peronet
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Salah Mecheri
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Artur Scherf
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Paola B Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| |
Collapse
|
20
|
Chellan P, Avery VM, Duffy S, Land KM, Tam CC, Kim JH, Cheng LW, Romero-Canelón I, Sadler PJ. Bioactive half-sandwich Rh and Ir bipyridyl complexes containing artemisinin. J Inorg Biochem 2021; 219:111408. [PMID: 33826972 DOI: 10.1016/j.jinorgbio.2021.111408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/21/2021] [Accepted: 02/21/2021] [Indexed: 02/06/2023]
Abstract
Reaction of dihydroartemisinin (DHA) with 4-methyl-4'-carboxy-2,2'-bipyridine yielded the new ester derivative L1. Six novel organometallic half-sandwich chlorido Rh(III) and Ir(III) complexes (1-6) containing pentamethylcyclopentadienyl, (Cp*), tetramethylphenylcyclopentadienyl (Cpxph), or tetramethylbiphenylcyclopentadienyl (Cpxbiph), and N,N-chelated bipyridyl group of L1, have been synthesized and characterized. The complexes were screened for inhibitory activity against the Plasmodium falciparum 3D7 (sensitive), Dd2 (multi-drug resistant) and NF54 late stage gametocytes (LSGNF54), the parasite strain Trichomonas vaginalis G3, as well as A2780 (human ovarian carcinoma), A549 (human alveolar adenocarcinoma), HCT116 (human colorectal carcinoma), MCF7 (human breast cancer) and PC3 (human prostate cancer) cancer cell lines. They show nanomolar antiplasmodial activity, outperforming chloroquine and artemisinin. Their activities were also comparable to dihydroartemisinin. As anticancer agents, several of the complexes showed high inhibitory effects, with Ir(III) complex 3, containing the tetramethylbiphenylcyclopentadienyl ligand, having similar IC50 values (concentration for 50% of maximum inhibition of cell growth) as the clinical drug cisplatin (1.06-9.23 μM versus 0.24-7.2 μM, respectively). Overall, the iridium complexes (1-3) are more potent compared to the rhodium derivatives (4-6), and complex 3 emerges as the most promising candidate for future studies.
Collapse
Affiliation(s)
- Prinessa Chellan
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Vicky M Avery
- Discovery Biology, Griffith University, Nathan, Queensland 4111, Australia
| | - Sandra Duffy
- Discovery Biology, Griffith University, Nathan, Queensland 4111, Australia
| | - Kirkwood M Land
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, United States of America
| | - Christina C Tam
- Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, United States of America
| | - Jong H Kim
- Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, United States of America
| | - Luisa W Cheng
- Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, United States of America
| | | | - Peter J Sadler
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
21
|
Nguyen W, Dans MG, Ngo A, Gancheva MR, Romeo O, Duffy S, de Koning-Ward TF, Lowes KN, Sabroux HJ, Avery VM, Wilson DW, Gilson PR, Sleebs BE. Structure activity refinement of phenylsulfonyl piperazines as antimalarials that block erythrocytic invasion. Eur J Med Chem 2021; 214:113253. [PMID: 33610028 DOI: 10.1016/j.ejmech.2021.113253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
The emerging resistance to combination therapies comprised of artemisinin derivatives has driven a need to identify new antimalarials with novel mechanisms of action. Central to the survival and proliferation of the malaria parasite is the invasion of red blood cells by Plasmodium merozoites, providing an attractive target for novel therapeutics. A screen of the Medicines for Malaria Venture Pathogen Box employing transgenic P. falciparum parasites expressing the nanoluciferase bioluminescent reporter identified the phenylsulfonyl piperazine class as a specific inhibitor of erythrocyte invasion. Here, we describe the optimization and further characterization of the phenylsulfonyl piperazine class. During the optimization process we defined the functionality required for P. falciparum asexual stage activity and determined the alpha-carbonyl S-methyl isomer was important for antimalarial potency. The optimized compounds also possessed comparable activity against multidrug resistant strains of P. falciparum and displayed weak activity against sexual stage gametocytes. We determined that the optimized compounds blocked erythrocyte invasion consistent with the asexual activity observed and therefore the phenylsulfonyl piperazine analogues described could serve as useful tools for studying Plasmodium erythrocyte invasion.
Collapse
Affiliation(s)
- William Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Madeline G Dans
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, 3004, Australia; School of Medicine, Deakin University, Waurn Ponds, 3216, Australia
| | - Anna Ngo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Maria R Gancheva
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Ornella Romeo
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Sandra Duffy
- Discovery Biology, Griffith University, Nathan, Queensland, 4111, Australia
| | | | - Kym N Lowes
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Helene Jousset Sabroux
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith University, Nathan, Queensland, 4111, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia; Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, 3004, Australia
| | - Paul R Gilson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, 3004, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
22
|
Investigation of factors affecting the production of P. falciparum gametocytes in an Indian isolate. 3 Biotech 2021; 11:55. [PMID: 33489674 DOI: 10.1007/s13205-020-02586-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022] Open
Abstract
The fundamental requirement of every gametocytocidal drug screening assay is the sufficient numbers of healthy and viable gametocytes. The number of in vitro gametocytes grossly depends on the genetic capacity of parasites to produce gametocytes and on various environmental factors that are not precisely elucidated. In the present study, we tested multiple environmental factors that are reported, hypothesized, or predicted to influence gametocyte numbers. We observed that hypoxanthine and the use of freshly drawn human blood significantly enhance gametocytemia (p < 0.05) in vitro. However, other tested factors did not significantly affect gametocytemia. The addition of N-acetyl glucosamine to the culture enriched the gametocytes but d-sorbitol (5% v/v) in amounts and duration of incubation tested was unable to do so without negatively affecting the maturity and health of the gametocytes. Although the in vitro gametocyte production depends on the genetic capability of the parasite strain tested, various environmental factors also control the ability of the strain to produce gametocytes up to a certain extent. This is the first study testing the role of various environmental factors that might affect the gametocyte development in a gametocyte producing strain. The results presented herein will help in the optimization of gametocyte production procedures for various gametocytocidal drug screening assays.
Collapse
|
23
|
Varela-Aramburu S, Ghosh C, Goerdeler F, Priegue P, Moscovitz O, Seeberger PH. Targeting and Inhibiting Plasmodium falciparum Using Ultra-small Gold Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2020; 12:43380-43387. [PMID: 32875786 PMCID: PMC7586288 DOI: 10.1021/acsami.0c09075] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/02/2020] [Indexed: 05/24/2023]
Abstract
Malaria, a mosquito-borne disease caused by Plasmodium species, claims more than 400,000 lives globally each year. The increasing drug resistance of the parasite renders the development of new anti-malaria drugs necessary. Alternatively, better delivery systems for already marketed drugs could help to solve the resistance problem. Herein, we report glucose-based ultra-small gold nanoparticles (Glc-NCs) that bind to cysteine-rich domains of Plasmodium falciparum surface proteins. Microscopy shows that Glc-NCs bind specifically to extracellular and all intra-erythrocytic stages of P. falciparum. Glc-NCs may be used as drug delivery agents as illustrated for ciprofloxacin, a poorly soluble antibiotic with low antimalarial activity. Ciprofloxacin conjugated to Glc-NCs is more water-soluble than the free drug and is more potent. Glyco-gold nanoparticles that target cysteine-rich domains on parasites may be helpful for the prevention and treatment of malaria.
Collapse
Affiliation(s)
- Silvia Varela-Aramburu
- Department of Biomolecular
Systems, Max Planck Institute of Colloids
and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Biology,
Chemistry, Pharmacy, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Chandradhish Ghosh
- Department of Biomolecular
Systems, Max Planck Institute of Colloids
and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Felix Goerdeler
- Department of Biomolecular
Systems, Max Planck Institute of Colloids
and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Biology,
Chemistry, Pharmacy, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Patricia Priegue
- Department of Biomolecular
Systems, Max Planck Institute of Colloids
and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Biology,
Chemistry, Pharmacy, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Oren Moscovitz
- Department of Biomolecular
Systems, Max Planck Institute of Colloids
and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Biology,
Chemistry, Pharmacy, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Peter H. Seeberger
- Department of Biomolecular
Systems, Max Planck Institute of Colloids
and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Biology,
Chemistry, Pharmacy, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| |
Collapse
|
24
|
Jennison C, Lucantoni L, O'Neill MT, McConville R, Erickson SM, Cowman AF, Sleebs BE, Avery VM, Boddey JA. Inhibition of Plasmepsin V Activity Blocks Plasmodium falciparum Gametocytogenesis and Transmission to Mosquitoes. Cell Rep 2020; 29:3796-3806.e4. [PMID: 31851913 DOI: 10.1016/j.celrep.2019.11.073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/14/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Plasmodium falciparum gametocytes infect mosquitoes and are responsible for malaria transmission. New interventions that block transmission could accelerate malaria elimination. Gametocytes develop within erythrocytes and activate protein export pathways that remodel the host cell. Plasmepsin V (PMV) is an aspartyl protease that is required for protein export in asexual parasites, but its function and essentiality in gametocytes has not been definitively proven, nor has PMV been assessed as a transmission-blocking drug target. Here, we show that PMV is expressed and can be inhibited specifically in P. falciparum stage I-II gametocytes. PMV inhibitors block processing and export of gametocyte effector proteins and inhibit development of stage II-V gametocytes. Gametocytogenesis in the presence of sublethal inhibitor concentrations results in stage V gametocytes that fail to infect mosquitoes. Therefore, PMV primes gametocyte effectors for export, which is essential for the development and fitness of gametocytes for transmission to mosquitoes.
Collapse
Affiliation(s)
- Charlie Jennison
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Leonardo Lucantoni
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan 4111, QLD, Australia
| | - Matthew T O'Neill
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia
| | - Robyn McConville
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Sara M Erickson
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan 4111, QLD, Australia
| | - Justin A Boddey
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia.
| |
Collapse
|
25
|
Tripathi AK, Mlambo G, Kanatani S, Sinnis P, Dimopoulos G. Plasmodium falciparum Gametocyte Culture and Mosquito Infection Through Artificial Membrane Feeding. J Vis Exp 2020. [PMID: 32716382 DOI: 10.3791/61426] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Malaria remains one of the most important public health problems, causing significant morbidity and mortality. Malaria is a mosquito borne disease transmitted through an infectious bite from the female Anopheles mosquito. Malaria control will eventually rely on a multitude of approaches, which includes ways to block transmission to, through and from mosquitoes. To study mosquito stages of malaria parasites in the laboratory, we have optimized a protocol to culture highly infectious Plasmodium falciparum gametocytes, a parasite stage required for transmission from the human host to the mosquito vector. P. falciparum gametocytes mature through five morphologically distinct steps, which takes approximately 1-2 weeks. Gametocyte culture described in this protocol is completed in 15 days and are infectious to mosquitoes from days 15-18. These protocols were developed to maintain a continuous cycle of infection competent gametocytes and to maintain uninterrupted supply of mosquito stages of the parasite. Here, we describe the methodology of gametocyte culture and how to infect mosquitoes with these parasites using glass membrane feeders.
Collapse
Affiliation(s)
- Abhai K Tripathi
- Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University;
| | - Godfree Mlambo
- Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University
| | - Sachie Kanatani
- Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University
| | - George Dimopoulos
- Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University
| |
Collapse
|
26
|
Dziekan JM, Wirjanata G, Dai L, Go KD, Yu H, Lim YT, Chen L, Wang LC, Puspita B, Prabhu N, Sobota RM, Nordlund P, Bozdech Z. Cellular thermal shift assay for the identification of drug-target interactions in the Plasmodium falciparum proteome. Nat Protoc 2020; 15:1881-1921. [PMID: 32341577 DOI: 10.1038/s41596-020-0310-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Despite decades of research, little is known about the cellular targets and the mode of action of the vast majority of antimalarial drugs. We recently demonstrated that the cellular thermal shift assay (CETSA) protocol in its two variants: the melt curve and the isothermal dose-response, represents a comprehensive strategy for the identification of antimalarial drug targets. CETSA enables proteome-wide target screening for unmodified antimalarial compounds with undetermined mechanisms of action, providing quantitative evidence about direct drug-protein interactions. The experimental workflow involves treatment of P. falciparum-infected erythrocytes with a compound of interest, heat exposure to denature proteins, soluble protein isolation, enzymatic digestion, peptide labeling with tandem mass tags, offline fractionation, and liquid chromatography-tandem mass spectrometry analysis. Methodological optimizations necessary for the analysis of this intracellular parasite are discussed, including enrichment of parasitized cells and hemoglobin depletion strategies to overcome high hemoglobin abundance in the host red blood cells. We outline an effective data processing workflow using the mineCETSA R package, which enables prioritization of drug-target candidates for follow-up studies. The entire protocol can be completed within 2 weeks.
Collapse
Affiliation(s)
- Jerzy Michal Dziekan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Grennady Wirjanata
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lingyun Dai
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Ka Diam Go
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Han Yu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yan Ting Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Liyan Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Loo Chien Wang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Brenda Puspita
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
27
|
Llorà-Batlle O, Michel-Todó L, Witmer K, Toda H, Fernández-Becerra C, Baum J, Cortés A. Conditional expression of PfAP2-G for controlled massive sexual conversion in Plasmodium falciparum. SCIENCE ADVANCES 2020; 6:eaaz5057. [PMID: 32577509 PMCID: PMC7286680 DOI: 10.1126/sciadv.aaz5057] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 04/15/2020] [Indexed: 05/19/2023]
Abstract
Malaria transmission requires that some asexual parasites convert into sexual forms termed gametocytes. The initial stages of sexual development, including sexually committed schizonts and sexual rings, remain poorly characterized, mainly because they are morphologically identical to their asexual counterparts and only a small subset of parasites undergo sexual development. Here, we describe a system for controlled sexual conversion in the human malaria parasite Plasmodium falciparum, based on conditional expression of the PfAP2-G transcription factor. Using this system, ~90 percent of the parasites converted into sexual forms upon induction, enabling the characterization of committed and early sexual stages without further purification. We characterized sexually committed schizonts and sexual rings at the transcriptomic and phenotypic levels, which revealed down-regulation of genes involved in solute transport upon sexual commitment, among other findings. The new inducible lines will facilitate the study of early sexual stages at additional levels, including multiomic characterization and drug susceptibility assays.
Collapse
Affiliation(s)
- Oriol Llorà-Batlle
- ISGlobal, Hospital Clinic–Universitat de Barcelona, Barcelona 08036, Catalonia, Spain
| | - Lucas Michel-Todó
- ISGlobal, Hospital Clinic–Universitat de Barcelona, Barcelona 08036, Catalonia, Spain
| | - Kathrin Witmer
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Haruka Toda
- ISGlobal, Hospital Clinic–Universitat de Barcelona, Barcelona 08036, Catalonia, Spain
| | - Carmen Fernández-Becerra
- ISGlobal, Hospital Clinic–Universitat de Barcelona, Barcelona 08036, Catalonia, Spain
- IGTP Institut d’Investigació Germans Trias i Pujol, Badalona 08916, Catalonia, Spain
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Alfred Cortés
- ISGlobal, Hospital Clinic–Universitat de Barcelona, Barcelona 08036, Catalonia, Spain
- ICREA, Barcelona 08010, Catalonia, Spain
- Corresponding author.
| |
Collapse
|
28
|
Abstract
As the world gets closer to eliminating malaria, the scientific community worldwide has begun to realize the importance of malaria transmission-blocking interventions. The onus of breaking the life cycle of the human malaria parasite Plasmodium falciparum predominantly rests upon transmission-blocking drugs because of emerging resistance to commonly used schizonticides and insecticides. This third part of our review series on malaria transmission-blocking entails transmission-blocking potential of preclinical transmission-blocking antimalarials and other non-malaria drugs/experimental compounds that are not in clinical or preclinical development for malaria but possess transmission-blocking potential. Collective analysis of the structure and the activity of these experimental compounds might pave the way toward generation of novel prototypes of next-generation transmission-blocking drugs.
Collapse
|
29
|
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
|
30
|
Nixon CP, Nixon CE, Michelow IC, Silva-Viera RA, Colantuono B, Obeidallah AS, Jha A, Dockery D, Raj D, Park S, Duffy PE, Kurtis JD. Antibodies to PfsEGXP, an Early Gametocyte-Enriched Phosphoprotein, Predict Decreased Plasmodium falciparum Gametocyte Density in Humans. J Infect Dis 2019; 218:1792-1801. [PMID: 29982707 DOI: 10.1093/infdis/jiy416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/03/2018] [Indexed: 11/14/2022] Open
Abstract
Background Antigametocyte-specific immune responses may regulate Plasmodium falciparum gametocyte density, providing the rationale for pursuing transmission-blocking vaccines (TBVs) that target gametocytes in the human host. Methods To identify novel antigametocyte TBV antigens, we interrogated the gametocyte proteome with our whole proteome differential screening method using plasma from a treatment-reinfection study conducted in western Kenya. At the start of the high-transmission season, 144 males (12-35 years) were enrolled and treated with quinine and doxycycline, peripheral venous blood samples were obtained, volunteers were observed, and weekly blood films were obtained for 18 weeks to quantify gametocytemia. Using plasma pooled from individuals with low versus high gametocyte carriage, we differentially screened a P falciparum gametocyte stage complementary deoxyribonucleic acid expression library. Results We identified 8 parasite genes uniquely recognized by gametocyte-resistant but not by gametocyte-susceptible individuals. Antibodies to one of these antigens, PfsEGXP, predicted lower gametocytemia measured over the 18-week transmission season (P = .021). When analyzed dichotomously, anti-PfsEGXP responders had 31% lower gametocyte density over 18 weeks of follow-up, compared with nonresponders (P = .04). Conclusions PfsEGXP is one of the first reported gametocyte-specific target of antibodies that predict decreased gametocyte density in humans and supports our novel TBV antigen discovery platform.
Collapse
Affiliation(s)
- Christian P Nixon
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence.,Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Christina E Nixon
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Ian C Michelow
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Rayna A Silva-Viera
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Bonnie Colantuono
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Aisha S Obeidallah
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Ambrish Jha
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Dominique Dockery
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Dipak Raj
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Sangshin Park
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Jonathan D Kurtis
- Center for International Health Research, Rhode Island Hospital and Alpert Medical School of Brown University, Providence.,Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| |
Collapse
|
31
|
van der Watt ME, Reader J, Churchyard A, Nondaba SH, Lauterbach SB, Niemand J, Abayomi S, van Biljon RA, Connacher JI, van Wyk RDJ, Le Manach C, Paquet T, González Cabrera D, Brunschwig C, Theron A, Lozano-Arias S, Rodrigues JFI, Herreros E, Leroy D, Duffy J, Street LJ, Chibale K, Mancama D, Coetzer TL, Birkholtz LM. Potent Plasmodium falciparum gametocytocidal compounds identified by exploring the kinase inhibitor chemical space for dual active antimalarials. J Antimicrob Chemother 2019; 73:1279-1290. [PMID: 29420756 DOI: 10.1093/jac/dky008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/02/2018] [Indexed: 01/06/2023] Open
Abstract
Objectives Novel chemical tools to eliminate malaria should ideally target both the asexual parasites and transmissible gametocytes. Several imidazopyridazines (IMPs) and 2-aminopyridines (2-APs) have been described as potent antimalarial candidates targeting lipid kinases. However, these have not been extensively explored for stage-specific inhibition of gametocytes in Plasmodium falciparum parasites. Here we provide an in-depth evaluation of the gametocytocidal activity of compounds from these chemotypes and identify novel starting points for dual-acting antimalarials. Methods We evaluated compounds against P. falciparum gametocytes using several assay platforms for cross-validation and stringently identified hits that were further profiled for stage specificity, speed of action and ex vivo efficacy. Physicochemical feature extraction and chemogenomic fingerprinting were applied to explore the kinase inhibition susceptibility profile. Results We identified 34 compounds with submicromolar activity against late stage gametocytes, validated across several assay platforms. Of these, 12 were potent at <100 nM (8 were IMPs and 4 were 2-APs) and were also active against early stage gametocytes and asexual parasites, with >1000-fold selectivity towards the parasite over mammalian cells. Front-runner compounds targeted mature gametocytes within 48 h and blocked transmission to mosquitoes. The resultant chemogenomic fingerprint of parasites treated with the lead compounds revealed the importance of targeting kinases in asexual parasites and gametocytes. Conclusions This study encompasses an in-depth evaluation of the kinase inhibitor space for gametocytocidal activity. Potent lead compounds have enticing dual activities and highlight the importance of targeting the kinase superfamily in malaria elimination strategies.
Collapse
Affiliation(s)
- Mariëtte E van der Watt
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Alisje Churchyard
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Sindisiwe H Nondaba
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Sonja B Lauterbach
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Jandeli Niemand
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Sijuade Abayomi
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Riëtte A van Biljon
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Jessica I Connacher
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Roelof D J van Wyk
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| | - Claire Le Manach
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Tanya Paquet
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Diego González Cabrera
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Christel Brunschwig
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Anjo Theron
- Biosciences, Council for Scientific and Industrial Research, PO Box 395, Pretoria 0001, South Africa
| | - Sonia Lozano-Arias
- GlaxoSmithKline, Tres Cantos Medicines Development Campus, Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain
| | - Janneth F I Rodrigues
- GlaxoSmithKline, Tres Cantos Medicines Development Campus, Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain
| | - Esperanza Herreros
- GlaxoSmithKline, Tres Cantos Medicines Development Campus, Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain
| | - Didier Leroy
- Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215, Geneva, Switzerland
| | - James Duffy
- Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215, Geneva, Switzerland
| | - Leslie J Street
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.,South African Medical Research Council, Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Dalu Mancama
- Biosciences, Council for Scientific and Industrial Research, PO Box 395, Pretoria 0001, South Africa
| | - Theresa L Coetzer
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Institute for Sustainable Malaria Control and South African Medical Research Council Collaborating Centre for Malaria Research, University of Pretoria, Private Bag x20, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
32
|
Yahiya S, Rueda-Zubiaurre A, Delves MJ, Fuchter MJ, Baum J. The antimalarial screening landscape-looking beyond the asexual blood stage. Curr Opin Chem Biol 2019; 50:1-9. [PMID: 30875617 PMCID: PMC6591700 DOI: 10.1016/j.cbpa.2019.01.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022]
Abstract
In recent years, the research agenda to tackle global morbidity and mortality from malaria disease has shifted towards innovation, in the hope that efforts at the frontiers of scientific research may re-invigorate gains made towards eradication. Discovery of new antimalarial drugs with novel chemotypes or modes of action lie at the heart of these efforts. There is a particular interest in drug candidates that target stages of the malaria parasite lifecycle beyond the symptomatic asexual blood stages. This is especially important given the spectre of emerging drug resistance to all current frontline antimalarials. One approach gaining increased interest is the potential of designing novel drugs that target parasite passage from infected individual to feeding mosquito and back again. Action of such therapeutics is geared much more at the population level rather than just concerned with the infected individual. The search for novel drugs active against these stages has been helped by improvements to in vitro culture of transmission and pre-erythrocytic parasite lifecycle stages, robotic automation and high content imaging, methodologies that permit the high-throughput screening (HTS) of compound libraries for drug discovery. Here, we review recent advances in the antimalarial screening landscape, focussed on transmission blocking as a key aim for drug-treatment campaigns of the future.
Collapse
Affiliation(s)
- Sabrina Yahiya
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Ainoa Rueda-Zubiaurre
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Michael J Delves
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Matthew J Fuchter
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK.
| |
Collapse
|
33
|
Pathak AK, Shiau JC, Thomas MB, Murdock CC. Cryogenically preserved RBCs support gametocytogenesis of Plasmodium falciparum in vitro and gametogenesis in mosquitoes. Malar J 2018; 17:457. [PMID: 30522507 PMCID: PMC6282341 DOI: 10.1186/s12936-018-2612-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/01/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The malaria Eradication Research Agenda (malERA) has identified human-to-mosquito transmission of Plasmodium falciparum as a major target for eradication. The cornerstone for identifying and evaluating transmission in the laboratory is standard membrane feeding assays (SMFAs) where mature gametocytes of P. falciparum generated in vitro are offered to mosquitoes as part of a blood-meal. However, propagation of "infectious" gametocytes requires 10-12 days with considerable physico-chemical demands imposed on host RBCs and thus, "fresh" RBCs that are ≤ 1-week old post-collection are generally recommended. However, in addition to the costs, physico-chemical characteristics unique to RBC donors may confound reproducibility and interpretation of SMFAs. Cryogenic storage of RBCs ("cryo-preserved RBCs") is accepted by European and US FDAs as an alternative to refrigeration (4 °C) for preserving RBC "quality" and while cryo-preserved RBCs have been used for in vitro cultures of other Plasmodia and the asexual stages of P. falciparum, none of the studies required RBCs to support parasite development for > 4 days. RESULTS Using the standard laboratory strain, P. falciparum NF54, 11 SMFAs were performed with RBCs from four separate donors to demonstrate that RBCs cryo-preserved in the gaseous phase of liquid nitrogen (- 196 °C) supported gametocytogenesis in vitro and subsequent gametogenesis in Anopheles stephensi mosquitoes. Overall levels of sporogony in the mosquito, as measured by oocyst and sporozoite prevalence, as well as oocyst burden, from each of the four donors thawed after varying intervals of cryopreservation (1, 4, 8, and 12 weeks) were comparable to using ≤ 1-week old refrigerated RBCs. Lastly, the potential for cryo-preserved RBCs to serve as a suitable alternative substrate is demonstrated for a Cambodian isolate of P. falciparum across two independent SMFAs. CONCLUSIONS Basic guidelines are presented for integrating cryo-preserved RBCs into an existing laboratory/insectary framework for P. falciparum SMFAs with significant potential for reducing running costs while achieving greater reliability. Lastly, scenarios are discussed where cryo-preserved RBCs may be especially useful in enhancing the understanding and/or providing novel insights into the patterns and processes underlying human-to-mosquito transmission.
Collapse
Affiliation(s)
- Ashutosh K Pathak
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA.
| | - Justine C Shiau
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Matthew B Thomas
- Center for Infectious Disease Dynamics and the Department of Entomology, Pennsylvania State University, State College, PA, 16803, USA
| | - Courtney C Murdock
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Odum School of Ecology, University of Georgia, Athens, GA, 30602, USA
- Center for Ecology of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA
- Center for Tropical Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, 30602, USA
- Riverbasin Center, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
34
|
Diedrich D, Stenzel K, Hesping E, Antonova-Koch Y, Gebru T, Duffy S, Fisher G, Schöler A, Meister S, Kurz T, Avery VM, Winzeler EA, Held J, Andrews KT, Hansen FK. One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites. Eur J Med Chem 2018; 158:801-813. [PMID: 30245402 PMCID: PMC6195125 DOI: 10.1016/j.ejmech.2018.09.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022]
Abstract
Malaria drug discovery has shifted from a focus on targeting asexual blood stage parasites, to the development of drugs that can also target exo-erythrocytic forms and/or gametocytes in order to prevent malaria and/or parasite transmission. In this work, we aimed to develop parasite-selective histone deacetylase inhibitors (HDACi) with activity against the disease-causing asexual blood stages of Plasmodium malaria parasites as well as with causal prophylactic and/or transmission blocking properties. An optimized one-pot, multi-component protocol via a sequential Ugi four-component reaction and hydroxylaminolysis was used for the preparation of a panel of peptoid-based HDACi. Several compounds displayed potent activity against drug-sensitive and drug-resistant P. falciparum asexual blood stages, high parasite-selectivity and submicromolar activity against exo-erythrocytic forms of P. berghei. Our optimization study resulted in the discovery of the hit compound 1u which combines high activity against asexual blood stage parasites (Pf 3D7 IC50: 4 nM; Pf Dd2 IC50: 1 nM) and P. berghei exo-erythrocytic forms (Pb EEF IC50: 25 nM) with promising parasite-specific activity (SIPf3D7/HepG2: 2496, SIPfDd2/HepG2: 9990, and SIPbEEF/HepG2: 400).
Collapse
Affiliation(s)
- Daniela Diedrich
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Katharina Stenzel
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany; Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Eva Hesping
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Yevgeniya Antonova-Koch
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Tamirat Gebru
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
| | - Sandra Duffy
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Gillian Fisher
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Andrea Schöler
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| | - Stephan Meister
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Thomas Kurz
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Vicky M Avery
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Jana Held
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
| | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia.
| | - Finn K Hansen
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany; Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany.
| |
Collapse
|
35
|
Duffy S, Avery VM. Routine In Vitro Culture of Plasmodium falciparum: Experimental Consequences? Trends Parasitol 2018; 34:564-575. [DOI: 10.1016/j.pt.2018.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
|
36
|
Kluska M, Komasińska M, Jabłońska J, Prukała W. Challenges of HPLC determination of quinoline derivatives used in the treatment of malaria. J LIQ CHROMATOGR R T 2018. [DOI: 10.1080/10826076.2018.1448870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Mariusz Kluska
- Faculty of Science, Siedlce University of Natural Sciences and Humanities, Institute of Chemistry, Siedlce, Poland
| | | | - Joanna Jabłońska
- Faculty of Science, Siedlce University of Natural Sciences and Humanities, Institute of Chemistry, Siedlce, Poland
| | - Wiesław Prukała
- Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
37
|
Duez J, Carucci M, Garcia-Barbazan I, Corral M, Perez O, Presa JL, Henry B, Roussel C, Ndour PA, Rosa NB, Sanz L, Gamo FJ, Buffet P. High-throughput microsphiltration to assess red blood cell deformability and screen for malaria transmission–blocking drugs. Nat Protoc 2018; 13:1362-1376. [DOI: 10.1038/nprot.2018.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Tougan T, Suzuki Y, Itagaki S, Izuka M, Toya Y, Uchihashi K, Horii T. An automated haematology analyzer XN-30 distinguishes developmental stages of falciparum malaria parasite cultured in vitro. Malar J 2018; 17:59. [PMID: 29391022 PMCID: PMC5796453 DOI: 10.1186/s12936-018-2208-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/27/2018] [Indexed: 01/31/2023] Open
Abstract
Background The automated haematology analyzer XN-30 (Sysmex, Kobe, Japan) easily and rapidly detects malarial parasites in clinical blood samples using flow cytometry. The XN-30 analyzer is able to distinguish each developmental stage by measuring DNA content and cell size. Thus, it was expected to be capable of quantifying the developmental stages of cultured falciparum parasite. To achieve this requirement, a modified algorithm was tested for its validity and reliability using in vitro cultured falciparum parasite. Results The XN-30 analyzer automatically measured each developmental stage as well as total parasitaemia. Comparison of the parasitaemia obtained using the XN-30 analyzer equipped with the modified algorithm with that obtained using microscopy examination of Giemsa-stained smears revealed the greater sensitivity and reproducibility of the former. The XN-30 analyzer also detected free merozoites and purified gametocytes. Conclusions The XN-30 analyzer allows the precise recognition and enumeration of total and each developmental stages of cultured falciparum parasites, and permits the sensitive and reproducible calculation of parasitaemia. The results indicate the potential of the XN-30 analyzer for basic research on malarial biology, anti-malarial drug discovery, and evaluation of drug efficacy. Electronic supplementary material The online version of this article (10.1186/s12936-018-2208-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takahiro Tougan
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuhgi Suzuki
- Sysmex Corporation, 4-4-4 Takatsukadai Nishiku, Kobe, Hyogo, 651-2271, Japan
| | - Sawako Itagaki
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Munehisa Izuka
- Sysmex Corporation, 4-4-4 Takatsukadai Nishiku, Kobe, Hyogo, 651-2271, Japan
| | - Yuji Toya
- Sysmex Corporation, 4-4-4 Takatsukadai Nishiku, Kobe, Hyogo, 651-2271, Japan
| | - Kinya Uchihashi
- Sysmex Corporation, 4-4-4 Takatsukadai Nishiku, Kobe, Hyogo, 651-2271, Japan
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
39
|
The malERA Refresh Consultative Panel on Basic Science and Enabling Technologies. malERA: An updated research agenda for basic science and enabling technologies in malaria elimination and eradication. PLoS Med 2017; 14:e1002451. [PMID: 29190277 PMCID: PMC5708601 DOI: 10.1371/journal.pmed.1002451] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Basic science holds enormous power for revealing the biological mechanisms of disease and, in turn, paving the way toward new, effective interventions. Recognizing this power, the 2011 Research Agenda for Malaria Eradication included key priorities in fundamental research that, if attained, could help accelerate progress toward disease elimination and eradication. The Malaria Eradication Research Agenda (malERA) Consultative Panel on Basic Science and Enabling Technologies reviewed the progress, continuing challenges, and major opportunities for future research. The recommendations come from a literature of published and unpublished materials and the deliberations of the malERA Refresh Consultative Panel. These areas span multiple aspects of the Plasmodium life cycle in both the human host and the Anopheles vector and include critical, unanswered questions about parasite transmission, human infection in the liver, asexual-stage biology, and malaria persistence. We believe an integrated approach encompassing human immunology, parasitology, and entomology, and harnessing new and emerging biomedical technologies offers the best path toward addressing these questions and, ultimately, lowering the worldwide burden of malaria.
Collapse
|
40
|
Stenzel K, Chua MJ, Duffy S, Antonova-Koch Y, Meister S, Hamacher A, Kassack MU, Winzeler E, Avery VM, Kurz T, Andrews KT, Hansen FK. Design and Synthesis of Terephthalic Acid-Based Histone Deacetylase Inhibitors with Dual-Stage Anti-Plasmodium Activity. ChemMedChem 2017; 12:1627-1636. [PMID: 28812327 DOI: 10.1002/cmdc.201700360] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/13/2017] [Indexed: 11/11/2022]
Abstract
In this work we aimed to develop parasite-selective histone deacetylase inhibitors (HDAC) inhibitors with activity against the disease-causing asexual blood stages of Plasmodium as well as causal prophylactic and/or transmission blocking properties. We report the design, synthesis, and biological testing of a series of 13 terephthalic acid-based HDAC inhibitors. All compounds showed low cytotoxicity against human embryonic kidney (HEK293) cells (IC50 : 8->51 μm), with 11 also having sub-micromolar in vitro activity against drug-sensitive (3D7) and multidrug-resistant (Dd2) asexual blood-stage P. falciparum parasites (IC50 ≈0.1-0.5 μm). A subset of compounds were examined for activity against early- and late-stage P. falciparum gametocytes and P. berghei exo-erythrocytic-stage parasites. While only moderate activity was observed against gametocytes (IC50 >2 μm), the most active compound (N1 -((3,5-dimethylbenzyl)oxy)-N4 -hydroxyterephthalamide, 1 f) showed sub-micromolar activity against P. berghei exo-erythrocytic stages (IC50 0.18 μm) and >270-fold better activity for exo-erythrocytic forms than for HepG2 cells. This, together with asexual-stage in vitro potency (IC50 ≈0.1 μm) and selectivity of this compound versus human cells (SI>450), suggests that 1 f may be a valuable starting point for the development of novel antimalarial drug leads with low host cell toxicity and multi-stage anti-plasmodial activity.
Collapse
Affiliation(s)
- Katharina Stenzel
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.,Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Ming Jang Chua
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Sandra Duffy
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Yevgeniya Antonova-Koch
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, USA
| | - Stephan Meister
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, USA
| | - Alexandra Hamacher
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Matthias U Kassack
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Elizabeth Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, USA
| | - Vicky M Avery
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Thomas Kurz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, QLD, 4111, Australia
| | - Finn K Hansen
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.,Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| |
Collapse
|
41
|
Screening the Medicines for Malaria Venture Pathogen Box across Multiple Pathogens Reclassifies Starting Points for Open-Source Drug Discovery. Antimicrob Agents Chemother 2017; 61:AAC.00379-17. [PMID: 28674055 PMCID: PMC5571359 DOI: 10.1128/aac.00379-17] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/19/2017] [Indexed: 01/19/2023] Open
Abstract
Open-access drug discovery provides a substantial resource for diseases primarily affecting the poor and disadvantaged. The open-access Pathogen Box collection is comprised of compounds with demonstrated biological activity against specific pathogenic organisms. The supply of this resource by the Medicines for Malaria Venture has the potential to provide new chemical starting points for a number of tropical and neglected diseases, through repurposing of these compounds for use in drug discovery campaigns for these additional pathogens. We tested the Pathogen Box against kinetoplastid parasites and malaria life cycle stages in vitro Consequently, chemical starting points for malaria, human African trypanosomiasis, Chagas disease, and leishmaniasis drug discovery efforts have been identified. Inclusive of this in vitro biological evaluation, outcomes from extensive literature reviews and database searches are provided. This information encompasses commercial availability, literature reference citations, other aliases and ChEMBL number with associated biological activity, where available. The release of this new data for the Pathogen Box collection into the public domain will aid the open-source model of drug discovery. Importantly, this will provide novel chemical starting points for drug discovery and target identification in tropical disease research.
Collapse
|
42
|
Leven M, Knaab TC, Held J, Duffy S, Meister S, Fischli C, Meitzner D, Lehmann U, Lungerich B, Kuna K, Stahlke P, Delves MJ, Buchholz M, Winzeler EA, Avery VM, Mordmüller B, Wittlin S, Kurz T. 3-Hydroxy-N'-arylidenepropanehydrazonamides with Halo-Substituted Phenanthrene Scaffolds Cure P. berghei Infected Mice When Administered Perorally. J Med Chem 2017; 60:6036-6044. [PMID: 28653845 DOI: 10.1021/acs.jmedchem.7b00140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Structural optimization of 3-hydroxy-N'-arylidenepropanehydrazonamides provided new analogs with nanomolar to subnanomolar antiplasmodial activity against asexual blood stages of Plasmodium falciparum, excellent parasite selectivity, and nanomolar activity against the earliest forms of gametocyte development. Particularly, derivatives with a 1,3-dihalo-6-trifluoromethylphenanthrene moiety showed outstanding in vivo properties and demonstrated in part curative activity in the Plasmodium berghei mouse model when administered perorally.
Collapse
Affiliation(s)
- Michael Leven
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Tanja C Knaab
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Jana Held
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen , Wilhelmstraße 27, 72074 Tübingen, Germany.,Centre de Recherches Medicales de Lambaréné , B.P.: 242 Lambaréné, Gabon
| | - Sandra Duffy
- Griffith Institute for Drug Discovery, Griffith University , Brisbane Innovation Park, Don Young Road, Nathan, Queensland 4111, Australia
| | - Stephan Meister
- Department of Pediatrics, School of Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Christoph Fischli
- Swiss Tropical and Public Health Institute , Socinstraße 57, 4002 Basel, Switzerland.,University of Basel , CH-4003 Basel, Switzerland
| | - Diane Meitzner
- Department of Drug Design and Target Validation, Fraunhofer-Institut für Zelltherapie und Immunologie , Weinbergweg 22, 06120 Halle (Saale), Germany
| | - Ursula Lehmann
- Swiss Tropical and Public Health Institute , Socinstraße 57, 4002 Basel, Switzerland.,University of Basel , CH-4003 Basel, Switzerland
| | - Beate Lungerich
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Krystina Kuna
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Petra Stahlke
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Michael J Delves
- Cell and Molecular Biology, Department of Life Sciences, Imperial College , London SW7 2AZ, U.K
| | - Mirko Buchholz
- Department of Drug Design and Target Validation, Fraunhofer-Institut für Zelltherapie und Immunologie , Weinbergweg 22, 06120 Halle (Saale), Germany
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Vicky M Avery
- Griffith Institute for Drug Discovery, Griffith University , Brisbane Innovation Park, Don Young Road, Nathan, Queensland 4111, Australia
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen , Wilhelmstraße 27, 72074 Tübingen, Germany.,Centre de Recherches Medicales de Lambaréné , B.P.: 242 Lambaréné, Gabon
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute , Socinstraße 57, 4002 Basel, Switzerland.,University of Basel , CH-4003 Basel, Switzerland
| | - Thomas Kurz
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
43
|
Duffy S, Avery VM. Plasmodium falciparum in vitro continuous culture conditions: A comparison of parasite susceptibility and tolerance to anti-malarial drugs throughout the asexual intra-erythrocytic life cycle. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2017; 7:295-302. [PMID: 28738214 PMCID: PMC5522918 DOI: 10.1016/j.ijpddr.2017.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 10/24/2022]
Abstract
The continuous culture of Plasmodium falciparum is often seen as a means to an end, that end being to probe the biology of the parasite in question, and ultimately for many in the malaria drug discovery arena, to identify means of killing the parasite in order to treat malaria. In vitro continuous culture of Plasmodium falciparum is a fundamental requirement when undertaking malaria research where the primary objectives utilise viable parasites of a desired lifecycle stage. This investigation, and resulting data, compared the impact culturing Plasmodium falciparum long term (4 months) in different environmental conditions had on experimental outcomes and thus conclusions. The example presented here focused specifically on the effect culture conditions had on the in vitro tolerance of Plasmodium falciparum to standard anti-malarial drugs, including artemisinin and lumefantrine. Historical data from an independent experiment for 3D7-ALB (5% O2) was also compared with that obtained from this study. We concluded that parasites cultured for several months in media supplemented with a serum substitute such as Albumax II® or within hyperoxic conditions (21% O2), demonstrate highly variable responses to artemisinin and lumefantrine but not all anti-malarial drugs, when compared to those cultured in human serum in combination with Albumax II® under normoxic conditions (5% O2) for the parasite.
Collapse
Affiliation(s)
- Sandra Duffy
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, 4111, Australia.
| | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, 4111, Australia
| |
Collapse
|
44
|
Demanga CG, Eng JWL, Gardiner DL, Roth A, Butterworth A, Adams JH, Trenholme KR, Dalton JP. The development of sexual stage malaria gametocytes in a Wave Bioreactor. Parasit Vectors 2017; 10:216. [PMID: 28464929 PMCID: PMC5414375 DOI: 10.1186/s13071-017-2155-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/25/2017] [Indexed: 11/14/2022] Open
Abstract
Background Blocking malaria gametocyte development in RBCs or their fertilization in the mosquito gut can prevent infection of the mosquito vector and passage of disease to the human host. A ‘transmission blocking’ strategy is a component of future malaria control. However, the lack of robust culture systems for producing large amounts of Plasmodium falciparum gametocytes has limited our understanding of sexual-stage malaria biology and made vaccine or chemotherapeutic discoveries more difficult. Methods The Wave BioreactorTM 20/50 EHT culture system was used to develop a convenient and low-maintenance protocol for inducing commitment of P. falciparum parasites to gametocytogenesis. Culture conditions were optimised to obtain mature stage V gametocytes within 2 weeks in a large-scale culture of up to a 1 l. Results We report a simple method for the induction of gametocytogenesis with N-acetylglucosamine (10 mM) within a Wave Bioreactor. By maintaining the culture for 14–16 days as many as 100 million gametocytes (stage V) were produced in a 1 l culture. Gametocytes isolated using magnetic activated cell sorting (MACS) columns were frozen in aliquots for storage. These were revitalised by thawing and shown to retain their ability to exflagellate and infect mosquitoes (Anopheles stephansi). Conclusions The production of gametocytes in the Wave Bioreactor under GMP-compliant conditions will not only facilitate cellular, developmental and molecular studies of gametocytes, but also the high-throughput screening for new anti-malarial drugs and, possibly, the development of whole-cell gametocyte or sporozoite-based vaccines. Electronic supplementary material The online version of this article (doi:10.1186/s13071-017-2155-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Corine G Demanga
- Institute of Parasitology, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Québec, H9X 3 V9, Canada
| | - Jenny W L Eng
- Institute of Parasitology, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Québec, H9X 3 V9, Canada
| | - Donald L Gardiner
- Malaria Biology Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, Brisbane, Australia.,School of Medicine, University of Queensland, St Lucia, 4072, QLD, Australia
| | - Alison Roth
- Department of Global Health, College of Public Health, University of South Florida, Tampa, 33612, FL, USA
| | - Alice Butterworth
- Malaria Biology Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, Brisbane, Australia
| | - John H Adams
- School of Biomolecular and Physical Sciences, Griffith University, Nathan, 4111, QLD, Australia
| | - Katharine R Trenholme
- Malaria Biology Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, Brisbane, Australia.,School of Biomolecular and Physical Sciences, Griffith University, Nathan, 4111, QLD, Australia
| | - John P Dalton
- Institute of Parasitology, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Québec, H9X 3 V9, Canada. .,School of Biological Sciences, Medical Biology Centre, Queen's University of Belfast, 97 Lisburn Road, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
45
|
The need to compare: assessing the level of agreement of three high-throughput assays against Plasmodium falciparum mature gametocytes. Sci Rep 2017; 7:45992. [PMID: 28378767 PMCID: PMC5380998 DOI: 10.1038/srep45992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022] Open
Abstract
Whole-cell High-Throughput Screening (HTS) is a key tool for the discovery of much needed malaria transmission blocking drugs. Discrepancies in the reported outcomes from various HTS Plasmodium falciparum gametocytocidal assays hinder the direct comparison of data and ultimately the interpretation of the transmission blocking potential of hits. To dissect the underlying determinants of such discrepancies and assess the impact that assay-specific factors have on transmission-blocking predictivity, a 39-compound subset from the Medicines for Malaria Venture Malaria Box was tested in parallel against three distinct mature stage gametocytocidal assays, under strictly controlled parasitological, chemical, temporal and analytical conditions resembling the standard membrane feeding assay (SMFA). Apart from a few assay-specific outliers, which highlighted the value of utilizing multiple complementary approaches, good agreement was observed (average ΔpIC50 of 0.12 ± 0.01). Longer compound incubation times improved the ability of the least sensitive assay to detect actives by 2-fold. Finally, combining the number of actives identified by any single assay with those obtained at longer incubation times yielded greatly improved outcomes and agreement with SMFA. Screening compounds using extended incubation times and using multiple in vitro assay technologies are valid approaches for the efficient identification of biologically relevant malaria transmission blocking hits.
Collapse
|
46
|
Pegoraro S, Duffey M, Otto TD, Wang Y, Rösemann R, Baumgartner R, Fehler SK, Lucantoni L, Avery VM, Moreno-Sabater A, Mazier D, Vial HJ, Strobl S, Sanchez CP, Lanzer M. SC83288 is a clinical development candidate for the treatment of severe malaria. Nat Commun 2017; 8:14193. [PMID: 28139658 PMCID: PMC5290327 DOI: 10.1038/ncomms14193] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 12/07/2016] [Indexed: 01/11/2023] Open
Abstract
Severe malaria is a life-threatening complication of an infection with the protozoan parasite Plasmodium falciparum, which requires immediate treatment. Safety and efficacy concerns with currently used drugs accentuate the need for new chemotherapeutic options against severe malaria. Here we describe a medicinal chemistry program starting from amicarbalide that led to two compounds with optimized pharmacological and antiparasitic properties. SC81458 and the clinical development candidate, SC83288, are fast-acting compounds that can cure a P. falciparum infection in a humanized NOD/SCID mouse model system. Detailed preclinical pharmacokinetic and toxicological studies reveal no observable drawbacks. Ultra-deep sequencing of resistant parasites identifies the sarco/endoplasmic reticulum Ca2+ transporting PfATP6 as a putative determinant of resistance to SC81458 and SC83288. Features, such as fast parasite killing, good safety margin, a potentially novel mode of action and a distinct chemotype support the clinical development of SC83288, as an intravenous application for the treatment of severe malaria.
Collapse
Affiliation(s)
| | - Maëlle Duffey
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Thomas D Otto
- Parasite Genomics, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Yulin Wang
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Roman Rösemann
- 4SC Discovery GmbH, Am Klopferspitz 19a, 82152 Martinsried, Germany
| | | | - Stefanie K Fehler
- 4SC AG, Am Klopferspitz 19a, 82152 Martinsried, Germany
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Leonardo Lucantoni
- Eskitis Institute for Drug Discovery, Griffith University, Don Young, Nathan Queensland 4111, Australia
| | - Vicky M Avery
- Eskitis Institute for Drug Discovery, Griffith University, Don Young, Nathan Queensland 4111, Australia
| | - Alicia Moreno-Sabater
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France
- AP-HP, Hôpital St Antoine, Service de Parasitologie-Mycologie, F-75012 Paris, France
| | - Dominique Mazier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France
- AP-HP, Groupe hospitalier La Pitié-Salpêtrière, Service de Parasitologie-Mycologie, F-75013 Paris, France
| | - Henri J Vial
- Dynamique des Interactions Membranaires Normales et Pathologiques, CNRS UMR 5235, Université Montpellier II, cc107, Place Eugène Bataillon, 34095 Montpellier, France
| | - Stefan Strobl
- 4SC Discovery GmbH, Am Klopferspitz 19a, 82152 Martinsried, Germany
| | - Cecilia P Sanchez
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Michael Lanzer
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
47
|
Gilson PR, Tan C, Jarman KE, Lowes KN, Curtis JM, Nguyen W, Di Rago AE, Bullen HE, Prinz B, Duffy S, Baell JB, Hutton CA, Jousset Subroux H, Crabb BS, Avery VM, Cowman AF, Sleebs BE. Optimization of 2-Anilino 4-Amino Substituted Quinazolines into Potent Antimalarial Agents with Oral in Vivo Activity. J Med Chem 2017; 60:1171-1188. [PMID: 28080063 DOI: 10.1021/acs.jmedchem.6b01673] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Novel antimalarial therapeutics that target multiple stages of the parasite lifecycle are urgently required to tackle the emerging problem of resistance with current drugs. Here, we describe the optimization of the 2-anilino quinazoline class as antimalarial agents. The class, identified from publicly available antimalarial screening data, was optimized to generate lead compounds that possess potent antimalarial activity against P. falciparum parasites comparable to the known antimalarials, chloroquine and mefloquine. During the optimization process, we defined the functionality necessary for activity and improved in vitro metabolism and solubility. The resultant lead compounds possess potent activity against a multidrug resistant strain of P. falciparum and arrest parasites at the ring phase of the asexual stage and also gametocytogensis. Finally, we show that the lead compounds are orally efficacious in a 4 day murine model of malaria disease burden.
Collapse
Affiliation(s)
- Paul R Gilson
- Macfarlane Burnet Institute for Medical Research and Public Health , Melbourne, 3004, Australia.,Monash University , Clayton, Victoria 3800, Australia
| | - Cyrus Tan
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| | - Kate E Jarman
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| | - Kym N Lowes
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| | - Joan M Curtis
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia
| | - William Nguyen
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| | - Adrian E Di Rago
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia
| | - Hayley E Bullen
- Macfarlane Burnet Institute for Medical Research and Public Health , Melbourne, 3004, Australia
| | - Boris Prinz
- Macfarlane Burnet Institute for Medical Research and Public Health , Melbourne, 3004, Australia
| | - Sandra Duffy
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University , Nathan, Queensland 4111, Australia
| | - Jonathan B Baell
- Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, Victoria 3052, Australia
| | - Craig A Hutton
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne , Parkville, Victoria 3010, Australia
| | - Helene Jousset Subroux
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| | - Brendan S Crabb
- Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia.,Macfarlane Burnet Institute for Medical Research and Public Health , Melbourne, 3004, Australia.,Monash University , Clayton, Victoria 3800, Australia
| | - Vicky M Avery
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University , Nathan, Queensland 4111, Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research , Parkville 3052, Australia.,Department of Medical Biology, The University of Melbourne , Parkville 3010, Australia
| |
Collapse
|
48
|
Fletcher S, Lucantoni L, Sykes ML, Jones AJ, Holleran JP, Saliba KJ, Avery VM. Biological characterization of chemically diverse compounds targeting the Plasmodium falciparum coenzyme A synthesis pathway. Parasit Vectors 2016; 9:589. [PMID: 27855724 PMCID: PMC5114727 DOI: 10.1186/s13071-016-1860-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/27/2016] [Indexed: 01/10/2023] Open
Abstract
Background In the fight against malaria, the discovery of chemical compounds with a novel mode of action and/or chemistry distinct from currently used drugs is vital to counteract the parasite’s known ability to develop drug resistance. Another desirable aspect is efficacy against gametocytes, the sexual developmental stage of the parasite which enables the transmission through Anopheles vectors. Using a chemical rescue approach, we previously identified compounds targeting Plasmodium falciparum coenzyme A (CoA) synthesis or utilization, a promising target that has not yet been exploited in anti-malarial drug development. Results We report on the outcomes of a series of biological tests that help to define the species- and stage-specificity, as well as the potential targets of these chemically diverse compounds. Compound activity against P. falciparum gametocytes was determined to assess stage-specificity and transmission-reducing potential. Against early stage gametocytes IC50 values ranging between 60 nM and 7.5 μM were obtained. With the exception of two compounds with sub-micromolar potencies across all intra-erythrocytic stages, activity against late stage gametocytes was lower. None of the compounds were specific pantothenate kinase inhibitors. Chemical rescue profiling with CoA pathway intermediates demonstrated that most compounds acted on either of the two final P. falciparum CoA synthesis enzymes, phosphopantetheine adenylyltransferase (PPAT) or dephospho CoA kinase (DPCK). The most active compound targeted either phosphopantothenoylcysteine synthetase (PPCS) or phosphopantothenoylcysteine decarboxylase (PPCDC). Species-specificity was evaluated against Trypanosoma cruzi and Trypanosoma brucei brucei. No specific activity against T. cruzi amastigotes was observed; however three compounds inhibited the viability of trypomastigotes with sub-micromolar potencies and were confirmed to act on T. b. brucei CoA synthesis. Conclusions Utilizing the compounds we previously identified as effective against asexual P. falciparum, we demonstrate for the first time that gametocytes, like the asexual stages, depend on CoA, with two compounds exhibiting sub-micromolar potencies across asexual forms and all gametocytes stages tested. Furthermore, three compounds inhibited the viability of T. cruzi and T. b. brucei trypomastigotes with sub-micromolar potencies and were confirmed to act on T. b. brucei CoA synthesis, indicating that the CoA synthesis pathway might represent a valuable new drug target in these parasite species. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1860-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sabine Fletcher
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Leonardo Lucantoni
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Melissa L Sykes
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Amy J Jones
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - John P Holleran
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Kevin J Saliba
- Medical School and Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Vicky M Avery
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|