1
|
Kalita T, Choudhury A, Shakya A, Ghosh SK, Singh UP, Bhat HR. A Review on Synthetic Thiazole Derivatives as an Antimalarial Agent. Curr Drug Discov Technol 2024; 21:e240124226141. [PMID: 38279721 DOI: 10.2174/0115701638276379231223101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Thiazole is a widely studied core structure in heterocyclic chemistry and has proven to be a valuable scaffold in medicinal chemistry. The presence of thiazole in both naturally occurring and synthetic pharmacologically active compounds demonstrates the adaptability of these derivatives. METHODS The current study attempted to review and compile the contributions of numerous researchers over the last 20 years to the medicinal importance of these scaffolds, with a primary focus on antimalarial activity. The review is based on an extensive search of PubMed, Google Scholar, Elsevier, and other renowned journal sites for a thorough literature survey involving various research and review articles. RESULTS A comprehensive review of the antimalarial activity of the thiazole scaffold revealed potential therapeutic targets in Plasmodium species. Furthermore, the correlation of structure-activity-relationship (SAR) studies from various articles suggests that the thiazole ring has therapeutic potential. CONCLUSION This article intends to point researchers in the right direction for developing potential thiazole-based compounds as antimalarial agents in the future.
Collapse
Affiliation(s)
- Tutumoni Kalita
- Department of Pharmaceutical Sciences, Girijananda Chowdhury Institute of Pharmaceutical Science, Hatkhowapara, Azara, Guwahati, Assam, India
| | - Ankita Choudhury
- Department of Pharmacy, Silchar Medical College and Hospital, Silchar, Assam, India
| | - Anshul Shakya
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Surajit Kumar Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Udaya Pratap Singh
- Drug Design & Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh, 211007, India
| | - Hans Raj Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| |
Collapse
|
2
|
Shirbhate E, Patel P, Patel VK, Veerasamy R, Sharma PC, Sinha BN, Rajak H. Synthetic and Semi-synthetic Drugs as a Promising Therapeutic Option for the Treatment of COVID-19. Mini Rev Med Chem 2021; 21:1004-1016. [PMID: 33280595 DOI: 10.2174/1389557520666201204162103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/23/2020] [Accepted: 10/16/2020] [Indexed: 11/22/2022]
Abstract
The novel coronavirus disease-19 (COVID-19) is a global pandemic that emerged from Wuhan, China, and has spread all around the world, affecting 216 countries or territories with 21,732,472 people infected and 770,866 deaths globally (as per WHO COVID-19 updates of August 18, 2020). Continuous efforts are being made to repurpose the existing drugs and develop vaccines for combating this infection. Despite, to date, no certified antiviral treatment or vaccine exists. Although, few candidates have displayed their efficacy in in vitro studies and are being repurposed for COVID- 19 treatment. This article summarizes synthetic and semi-synthetic compounds displaying potent activity in clinical uses or studies on COVID-19 and also focuses on the mode of action of drugs being repositioned against COVID-19.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur 495 009, C.G., India
| | - Preeti Patel
- Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur 495 009, C.G., India
| | - Vijay K Patel
- Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur 495 009, C.G., India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Prabodh C Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra 136 119, Haryana, India
| | - Barij N Sinha
- Department of Pharmacy, Birla Institute of Technology, Mesra - Ranchi-835 215, Jharkhand, India
| | - Harish Rajak
- Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur 495 009, C.G., India
| |
Collapse
|
3
|
Barber J, Sikakana P, Sadler C, Baud D, Valentin JP, Roberts R. A target safety assessment of the potential toxicological risks of targeting plasmepsin IX/X for the treatment of malaria. Toxicol Res (Camb) 2021; 10:203-213. [PMID: 33884171 DOI: 10.1093/toxres/tfaa106] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
The aspartic proteases plasmepsin IX/X are important antimalarial drug targets due to their specificity to the malaria parasite and their vital role as mediators of disease progression. Focusing on parasite-specific targets where no human homologue exists reduces the possibility of on-target drug toxicity. However, there is a risk of toxicity driven by inadequate selectivity for plasmepsins IX/X in Plasmodium over related mammalian aspartic proteases. Of these, CatD/E may be of most toxicological relevance as CatD is a ubiquitous lysosomal enzyme present in most cell types and CatE is found in the gut and in erythrocytes, the clinically significant site of malarial infection. Based on mammalian aspartic protease physiology and adverse drug reactions (ADRs) to FDA-approved human immunodeficiency virus (HIV) aspartic protease inhibitors, we predicted several potential toxicities including β-cell and congenital abnormalities, hypotension, hypopigmentation, hyperlipidaemia, increased infection risk and respiratory, renal, gastrointestinal, dermatological, and other epithelial tissue toxicities. These ADRs to the HIV treatments are likely to be a result of host aspartic protease inhibition due a lack of specificity for the HIV protease; plasmepsins are much more closely related to human CatD than to HIV proteinase. Plasmepsin IX/X inhibition presents an opportunity to specifically target Plasmodium as an effective antimalarial treatment, providing adequate selectivity can be obtained. Potential plasmepsin IX/X inhibitors should be assayed for inhibitory activity against the main human aspartic proteases and particularly CatD/E. An investigative rodent study conducted early in drug discovery would serve as an initial risk assessment of the potential hazards identified.
Collapse
Affiliation(s)
- Jane Barber
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| | | | | | - Delphine Baud
- Medicines for Malaria Venture, 20 Route de Pré-Bois, Geneva 1215, Switzerland
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Building R9, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Ruth Roberts
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| |
Collapse
|
4
|
Ansbro MR, Itkin Z, Chen L, Zahoranszky-Kohalmi G, Amaratunga C, Miotto O, Peryea T, Hobbs CV, Suon S, Sá JM, Dondorp AM, van der Pluijm RW, Wellems TE, Simeonov A, Eastman RT. Modulation of Triple Artemisinin-Based Combination Therapy Pharmacodynamics by Plasmodium falciparum Genotype. ACS Pharmacol Transl Sci 2020; 3:1144-1157. [PMID: 33344893 PMCID: PMC7737215 DOI: 10.1021/acsptsci.0c00110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 01/19/2023]
Abstract
The first-line treatments for uncomplicated Plasmodium falciparum malaria are artemisinin-based combination therapies (ACTs), consisting of an artemisinin derivative combined with a longer acting partner drug. However, the spread of P. falciparum with decreased susceptibility to artemisinin and partner drugs presents a significant challenge to malaria control efforts. To stem the spread of drug resistant parasites, novel chemotherapeutic strategies are being evaluated, including the implementation of triple artemisinin-based combination therapies (TACTs). Currently, there is limited knowledge on the pharmacodynamic and pharmacogenetic interactions of proposed TACT drug combinations. To evaluate these interactions, we established an in vitro high-throughput process for measuring the drug concentration-response to three distinct antimalarial drugs present in a TACT. Sixteen different TACT combinations were screened against 15 parasite lines from Cambodia, with a focus on parasites with differential susceptibilities to piperaquine and artemisinins. Analysis revealed drug-drug interactions unique to specific genetic backgrounds, including antagonism between piperaquine and pyronaridine associated with gene amplification of plasmepsin II/III, two aspartic proteases that localize to the parasite digestive vacuole. From this initial study, we identified parasite genotypes with decreased susceptibility to specific TACTs, as well as potential TACTs that display antagonism in a genotype-dependent manner. Our assay and analysis platform can be further leveraged to inform drug implementation decisions and evaluate next-generation TACTs.
Collapse
Affiliation(s)
- Megan R. Ansbro
- Laboratory of Malaria
and Vector Research, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
- Wellcome Sanger Institute, Hinxton CB10 1SA, U.K.
| | - Zina Itkin
- National
Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Lu Chen
- National
Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Gergely Zahoranszky-Kohalmi
- National
Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Chanaki Amaratunga
- Laboratory of Malaria
and Vector Research, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Olivo Miotto
- Wellcome Sanger Institute, Hinxton CB10 1SA, U.K.
- Mahidol-Oxford Tropical Medicine Research
Unit, Faculty of Tropical Medicine, Mahidol
University, Bangkok 10400, Thailand
- Centre
for Tropical Medicine and Global Health, Nuffield Department of Medicine
Research, University of Oxford, Oxford OX3 7LF, U.K.
- Medical Research Council (MRC) Centre for Genomics and
Global Health, University of Oxford, Oxford OX3 7BN, U.K.
| | - Tyler Peryea
- National
Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Charlotte V. Hobbs
- Division of Infectious Diseases, Children’s
Hospital, University of Mississippi Medical
Center, Jackson, Mississippi 39216, United States
| | - Seila Suon
- National Center for Parasitology, Entomology,
and Malaria Control, Phnom Penh, Cambodia
| | - Juliana M. Sá
- Laboratory of Malaria
and Vector Research, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Arjen M. Dondorp
- Mahidol-Oxford Tropical Medicine Research
Unit, Faculty of Tropical Medicine, Mahidol
University, Bangkok 10400, Thailand
- Centre
for Tropical Medicine and Global Health, Nuffield Department of Medicine
Research, University of Oxford, Oxford OX3 7LF, U.K.
| | - Rob W. van der Pluijm
- Mahidol-Oxford Tropical Medicine Research
Unit, Faculty of Tropical Medicine, Mahidol
University, Bangkok 10400, Thailand
- Centre
for Tropical Medicine and Global Health, Nuffield Department of Medicine
Research, University of Oxford, Oxford OX3 7LF, U.K.
| | - Thomas E. Wellems
- Laboratory of Malaria
and Vector Research, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Anton Simeonov
- National
Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Richard T. Eastman
- National
Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| |
Collapse
|
5
|
Azevedo R, Mendes AM, Prudêncio M. The Impact of Antiretroviral Therapy on Malaria Parasite Transmission. Front Microbiol 2020; 10:3048. [PMID: 32038528 PMCID: PMC6993566 DOI: 10.3389/fmicb.2019.03048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Coendemicity between the human immunodeficiency virus (HIV) and Plasmodium parasites, the causative agents of acquired immunodeficiency syndrome (AIDS) and malaria, respectively, occurs in several regions around the world. Although the impact of the interaction between these two organisms is not well understood, it is thought that the outcome of either disease may be negatively influenced by coinfection. Therefore, it is important to understand how current first-line antiretroviral therapies (ART) might impact Plasmodium infection in these regions. Here, we describe the effect of 18 antiretroviral compounds and of first-line ART on the blood and sporogonic stages of Plasmodium berghei in vitro and in vivo. We show that the combination zidovudine + lamivudine + lopinavir/ritonavir (LPV/r), employed as first-line HIV treatment in the field, has a strong inhibitory activity on the sporogonic stages of P. berghei and that several non-nucleoside reverse transcriptase inhibitors (NNRTI) have a moderate effect on this stage of the parasite’s life cycle. Our results expose the effect of current first-line ART on Plasmodium infection and identify potential alternative therapies for HIV/AIDS that might impact malaria transmission.
Collapse
Affiliation(s)
- Raquel Azevedo
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - António M Mendes
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Miguel Prudêncio
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
6
|
Skinner-Adams TS, Fisher GM, Riches AG, Hutt OE, Jarvis KE, Wilson T, von Itzstein M, Chopra P, Antonova-Koch Y, Meister S, Winzeler EA, Clarke M, Fidock DA, Burrows JN, Ryan JH, Andrews KT. Cyclization-blocked proguanil as a strategy to improve the antimalarial activity of atovaquone. Commun Biol 2019; 2:166. [PMID: 31069275 PMCID: PMC6499835 DOI: 10.1038/s42003-019-0397-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/15/2019] [Indexed: 12/28/2022] Open
Abstract
Atovaquone-proguanil (Malarone®) is used for malaria prophylaxis and treatment. While the cytochrome bc1-inhibitor atovaquone has potent activity, proguanil's action is attributed to its cyclization-metabolite, cycloguanil. Evidence suggests that proguanil has limited intrinsic activity, associated with mitochondrial-function. Here we demonstrate that proguanil, and cyclization-blocked analogue tBuPG, have potent, but slow-acting, in vitro anti-plasmodial activity. Activity is folate-metabolism and isoprenoid biosynthesis-independent. In yeast dihydroorotate dehydrogenase-expressing parasites, proguanil and tBuPG slow-action remains, while bc1-inhibitor activity switches from comparatively fast to slow-acting. Like proguanil, tBuPG has activity against P. berghei liver-stage parasites. Both analogues act synergistically with bc1-inhibitors against blood-stages in vitro, however cycloguanil antagonizes activity. Together, these data suggest that proguanil is a potent slow-acting anti-plasmodial agent, that bc1 is essential to parasite survival independent of dihydroorotate dehydrogenase-activity, that Malarone® is a triple-drug combination that includes antagonistic partners and that a cyclization-blocked proguanil may be a superior combination partner for bc1-inhibitors in vivo.
Collapse
Affiliation(s)
- Tina S. Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111 Australia
| | - Gillian M. Fisher
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111 Australia
| | - Andrew G. Riches
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, VIC 3168 Australia
| | - Oliver E. Hutt
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, VIC 3168 Australia
| | - Karen E. Jarvis
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, VIC 3168 Australia
| | - Tony Wilson
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, VIC 3168 Australia
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University Gold Coast Campus, Gold Coast, QLD 4222 Australia
| | - Pradeep Chopra
- Institute for Glycomics, Griffith University Gold Coast Campus, Gold Coast, QLD 4222 Australia
| | - Yevgeniya Antonova-Koch
- School of Medicine, University of California, San Diego, La Jolla, CA 92093 USA
- Present Address: California Institute for Biomedical Research (Calibr), La Jolla, CA 92037 USA
| | - Stephan Meister
- School of Medicine, University of California, San Diego, La Jolla, CA 92093 USA
- Present Address: Beckman Coulter Life Sciences in Indianapolis, Indianapolis, IN 46268 USA
| | | | - Mary Clarke
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111 Australia
| | - David A. Fidock
- Department of Microbiology and Immunology, and Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032 USA
| | - Jeremy N. Burrows
- Medicines for Malaria Venture (MMV), Route de Pré Bois 20, Geneva, 1215 Switzerland
| | - John H. Ryan
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, VIC 3168 Australia
| | - Katherine T. Andrews
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111 Australia
| |
Collapse
|
7
|
Kumar S, Bhardwaj TR, Prasad DN, Singh RK. Drug targets for resistant malaria: Historic to future perspectives. Biomed Pharmacother 2018; 104:8-27. [PMID: 29758416 DOI: 10.1016/j.biopha.2018.05.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/22/2018] [Accepted: 05/07/2018] [Indexed: 01/05/2023] Open
Abstract
New antimalarial targets are the prime need for the discovery of potent drug candidates. In order to fulfill this objective, antimalarial drug researches are focusing on promising targets in order to develop new drug candidates. Basic metabolism and biochemical process in the malaria parasite, i.e. Plasmodium falciparum can play an indispensable role in the identification of these targets. But, the emergence of resistance to antimalarial drugs is an escalating comprehensive problem with the progress of antimalarial drug development. The development of resistance has highlighted the need for the search of novel antimalarial molecules. The pharmaceutical industries are committed to new drug development due to the global recognition of this life threatening resistance to the currently available antimalarial therapy. The recent developments in the understanding of parasite biology are exhilarating this resistance issue which is further being ignited by malaria genome project. With this background of information, this review was aimed to highlights and provides useful information on various present and promising treatment approaches for resistant malaria, new progresses, pursued by some innovative targets that have been explored till date. This review also discusses modern and futuristic multiple approaches to antimalarial drug discovery and development with pictorial presentations highlighting the various targets, that could be exploited for generating promising new drugs in the future for drug resistant malaria.
Collapse
Affiliation(s)
- Sahil Kumar
- School of Pharmacy and Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi, Dist. Solan, 173205, Himachal Pradesh, India
| | - T R Bhardwaj
- School of Pharmacy and Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi, Dist. Solan, 173205, Himachal Pradesh, India
| | - D N Prasad
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India
| | - Rajesh K Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India.
| |
Collapse
|
8
|
Purification and antiparasitic activity of a few legume serine proteinase inhibitors: Effect on erythrocyte invasion, schizont rupture and proteolytic processing of the Plasmodium falciparum AMA1 protein. Process Biochem 2017. [DOI: 10.1016/j.procbio.2017.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
9
|
Synthesis, biological characterisation and structure activity relationships of aromatic bisamidines active against Plasmodium falciparum. Eur J Med Chem 2017; 127:22-40. [DOI: 10.1016/j.ejmech.2016.12.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 01/27/2023]
|
10
|
Chua MJ, Arnold MSJ, Xu W, Lancelot J, Lamotte S, Späth GF, Prina E, Pierce RJ, Fairlie DP, Skinner-Adams TS, Andrews KT. Effect of clinically approved HDAC inhibitors on Plasmodium, Leishmania and Schistosoma parasite growth. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2016; 7:42-50. [PMID: 28107750 PMCID: PMC5241585 DOI: 10.1016/j.ijpddr.2016.12.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/11/2022]
Abstract
Malaria, schistosomiasis and leishmaniases are among the most prevalent tropical parasitic diseases and each requires new innovative treatments. Targeting essential parasite pathways, such as those that regulate gene expression and cell cycle progression, is a key strategy for discovering new drug leads. In this study, four clinically approved anti-cancer drugs (Vorinostat, Belinostat, Panobinostat and Romidepsin) that target histone/lysine deacetylase enzymes were examined for in vitro activity against Plasmodium knowlesi, Schistosoma mansoni, Leishmania amazonensis and L. donovani parasites and two for in vivo activity in a mouse malaria model. All four compounds were potent inhibitors of P. knowlesi malaria parasites (IC50 9-370 nM), with belinostat, panobinostat and vorinostat having 8-45 fold selectivity for the parasite over human neonatal foreskin fibroblast (NFF) or human embryonic kidney (HEK 293) cells, while romidepsin was not selective. Each of the HDAC inhibitor drugs caused hyperacetylation of P. knowlesi histone H4. None of the drugs was active against Leishmania amastigote or promastigote parasites (IC50 > 20 μM) or S. mansoni schistosomula (IC50 > 10 μM), however romidepsin inhibited S. mansoni adult worm parings and egg production (IC50 ∼10 μM). Modest in vivo activity was observed in P. berghei infected mice dosed orally with vorinostat or panobinostat (25 mg/kg twice daily for four days), with a significant reduction in parasitemia observed on days 4-7 and 4-10 after infection (P < 0.05), respectively.
Collapse
Affiliation(s)
- Ming Jang Chua
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Megan S J Arnold
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Weijun Xu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Julien Lancelot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204- CIIL -Centre D'Infection et D'Immunité de Lille, F-59000 Lille, France
| | - Suzanne Lamotte
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Gerald F Späth
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Eric Prina
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Raymond J Pierce
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204- CIIL -Centre D'Infection et D'Immunité de Lille, F-59000 Lille, France
| | - David P Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Tina S Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia.
| |
Collapse
|
11
|
Abiodun OO, Gbimadee N, Gbotosho GO. Lopinavir/ritonavir enhanced the antimalarial activity of amodiaquine and artesunate in a mouse model of Plasmodium berghei. J Chemother 2016; 28:482-486. [PMID: 26900802 DOI: 10.1080/1120009x.2016.1139770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Treatment of malaria and HIV in co-infected patients remains a challenge due to the limited information on interaction between drugs used for the treatment of the two infections. Thus, this study evaluated the interaction between lopinavir/ritonavir (LR) and artesunate (AS), amodiaquine (AQ) or a fixed dose of AS/AQ in a mouse model of chloroquine-resistant Plasmodium berghei. Combination of LR with graded doses of AS or AQ resulted in a significant reduced ED50. In addition, parasites cleared completely from day 3 till day 21 post-infection in animals infected, treated with AS/AQ alone or AS/AQ with LR and all the animals survived till day 21 post-infection. In contrast, survival on day 21 in animals treated with AQ alone or AQ with LR was 20 and 60%, respectively. It appears that the protease inhibitor LR enhanced the antimalarial drugs AS and AQ.
Collapse
Affiliation(s)
| | - Nekabari Gbimadee
- a Department of Pharmacology and Therapeutics , College of Medicine, University of Ibadan , Ibadan , Nigeria
| | - Grace Olushola Gbotosho
- a Department of Pharmacology and Therapeutics , College of Medicine, University of Ibadan , Ibadan , Nigeria
| |
Collapse
|
12
|
Skinner-Adams TS, Sumanadasa SD, Fisher GM, Davis RA, Doolan DL, Andrews KT. Defining the targets of antiparasitic compounds. Drug Discov Today 2016; 21:725-39. [DOI: 10.1016/j.drudis.2016.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/04/2015] [Accepted: 01/07/2016] [Indexed: 10/22/2022]
|
13
|
Liu P, Robbins AH, Marzahn MR, McClung SH, Yowell CA, Stevens SM, Dame JB, Dunn BM. Enzymatic Characterization of Recombinant Food Vacuole Plasmepsin 4 from the Rodent Malaria Parasite Plasmodium berghei. PLoS One 2015; 10:e0141758. [PMID: 26510189 PMCID: PMC4624963 DOI: 10.1371/journal.pone.0141758] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/12/2015] [Indexed: 01/17/2023] Open
Abstract
The rodent malaria parasite Plasmodium berghei is a practical model organism for experimental studies of human malaria. Plasmepsins are a class of aspartic proteinase isoforms that exert multiple pathological effects in malaria parasites. Plasmepsins residing in the food vacuole (FV) of the parasite hydrolyze hemoglobin in red blood cells. In this study, we cloned PbPM4, the FV plasmepsin gene of P. berghei that encoded an N-terminally truncated pro-segment and the mature enzyme from genomic DNA. We over-expressed this PbPM4 zymogen as inclusion bodies (IB) in Escherichia coli, and purified the protein following in vitro IB refolding. Auto-maturation of the PbPM4 zymogen to mature enzyme was carried out at pH 4.5, 5.0, and 5.5. Interestingly, we found that the PbPM4 zymogen exhibited catalytic activity regardless of the presence of the pro-segment. We determined the optimal catalytic conditions for PbPM4 and studied enzyme kinetics on substrates and inhibitors of aspartic proteinases. Using combinatorial chemistry-based peptide libraries, we studied the active site preferences of PbPM4 at subsites S1, S2, S3, S1’, S2’ and S3’. Based on these results, we designed and synthesized a selective peptidomimetic compound and tested its inhibition of PbPM4, seven FV plasmepsins from human malaria parasites, and human cathepsin D (hcatD). We showed that this compound exhibited a >10-fold selectivity to PbPM4 and human malaria parasite plasmepsin 4 orthologs versus hcatD. Data from this study furthesr our understanding of enzymatic characteristics of the plasmepsin family and provides leads for anti-malarial drug design.
Collapse
Affiliation(s)
- Peng Liu
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
- * E-mail: (PL); (BMD)
| | - Arthur H. Robbins
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - Melissa R. Marzahn
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - Scott H. McClung
- Protein Core, Interdisciplinary Center for Biotechnology Research, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - Charles A. Yowell
- Department of Infectious Diseases and Pathology, University of Florida, College of Veterinary Medicine, Gainesville, Florida, United States of America
| | - Stanley M. Stevens
- Protein Core, Interdisciplinary Center for Biotechnology Research, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - John B. Dame
- Department of Infectious Diseases and Pathology, University of Florida, College of Veterinary Medicine, Gainesville, Florida, United States of America
| | - Ben M. Dunn
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
- * E-mail: (PL); (BMD)
| |
Collapse
|
14
|
Pharmacokinetic interactions between artesunate-mefloquine and ritonavir-boosted lopinavir in healthy Thai adults. Malar J 2015; 14:400. [PMID: 26452725 PMCID: PMC4600319 DOI: 10.1186/s12936-015-0916-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/25/2015] [Indexed: 11/30/2022] Open
Abstract
Background Concomitant use of anti-malarial and antiretroviral drugs is increasingly frequent in malaria and HIV endemic regions. The aim of the study was to investigate the pharmacokinetic interaction between the anti-malarial drugs, artesunate-mefloquine and the antiretroviral drug, lopinavir boosted with ritonavir (LPV/r). Methods The study was an open-label, three-way, sequential, cross-over, pharmacokinetic study in healthy Thai adults. Subjects received the following treatments: Period 1: standard 3-day artesunate-mefloquine combination; Period 2 (2 months wash-out): oral LPV/r 400 mg/100 mg twice a day for 14 days; and, Period 3: artesunate-mefloquine and LPV/r twice a day for 3 days. Sixteen subjects (eight females) were enrolled and pharmacokinetic parameters were determined by non-compartmental analysis. Results In the presence of LPV/r, artesunate Cmax and systemic exposure were significantly increased by 45–80 %, while the metabolic ratio of dihydroartemisinin to artesunate was significantly reduced by 72 %. In addition, mefloquine Cmax and systemic exposure were significantly reduced by 19–37 %. In the presence of artesunate-mefloquine, lopinavir Cmax was significantly reduced by 22 % but without significant change in systemic drug exposure. The 90 % CI of the geometric mean ratio (GMR) of AUC0−∞ and Cmax were outside the acceptable bioequivalent range for each drug. Drug treatments were generally well tolerated with no serious adverse events. Vertigo, nausea and vomiting were the most common adverse events reported. Conclusion The reduction in systemic exposure of all investigated drugs raises concerns of an increased risk of treatment failure rate in co-infected patients and should be further investigated.
Collapse
|
15
|
Rainsford KD, Parke AL, Clifford-Rashotte M, Kean WF. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology 2015; 23:231-69. [PMID: 26246395 DOI: 10.1007/s10787-015-0239-y] [Citation(s) in RCA: 349] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/23/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVES This review examines the pharmacokinetics, modes of action and therapeutic properties of the anti-malarial drugs, hydroxychloroquine (HCQ) and chloroquine (CQ), in the treatment of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and related conditions, as well as osteoarthritis (OA). KEY FINDINGS Both HCQ and CQ have historically been employed successfully for the treatment of SLE and RA for over 70 years. HCQ has been used extensively for SLE where it has a good reputation for controlling the dermatological complications in SLE. It has also been reported to effectively control the symptoms of Sjøgren's syndrome, as well as preventing thrombosis in phospholipid antibody (aPL) syndrome. In RA and SLE, HCQ is preferred because of the lower incidence of gastrointestinal adverse reactions compared with CQ and it might have a lower risk of ocular adverse reactions. There is increasing evidence that HCQ may reduce atherosclerosis and risks of cardiovascular disease in rheumatic patients. Both HCQ and CQ have been shown to improve glycaemia and reduce the risks of type II diabetes mellitus. Although both HCQ and CQ are effective in low-moderate RA, HCQ is now preferred as part of combination therapy for more severe disease. The advantages of combination therapy are that the doses of the individual drugs may be lowered so reducing adverse reactions. Both HCQ and CQ are diastereoisomers, have basic properties and are given as the sulphate and phosphate salts. While being relatively well absorbed orally and with good bioavailability, they have long and variable plasma terminal elimination half-lives (approximately 40-60 days). This reflects their high volume of distribution, V D (HCQ 44,000L; CQ 65,000L) which extends into aqueous compartments, long mean residence time (HCQ 1300 h; CQ 900 h) and with about half the drugs (metabolites) undergoing renal clearance. The strong binding to melanin reflects the ocular injury and dermatological properties of these drugs. The consensus is that the occurrence of ocular adverse reactions can be minimised by close attention to the dose (which should be set on a body weight basis) with regular (e.g. quarterly) retinal examination. Although HCQ and CQ can pass through the placenta, the use of these drugs during pregnancy does not appear to risk harm to the baby and might be beneficial to the mother with SLE and her child by controlling the SLE disease activity, which is known to be an important factor affecting pregnancy outcome. The modes of action of HCQ and CQ in these arthritides represent somewhat of an enigma. Undoubtedly, these drugs have multiple actions related, in part, their ability to accumulate in lysosomes and autophagosomes of phagocytic cells as well as affecting MHC Class II expression and antigen presentation; actions of the production of pro-inflammatory cytokines [e.g. interleukin-1 (IL-1) tumour necrosis factor-α (TNFα)]; control of toll-like receptor-9 activation; and leucocyte generation of reactive oxygen species (ROS); i.e. antioxidant activity. The actions of these drugs on T and B cells are less clear but may depend on these leucocyte-mediated actions. Anti-malarials also protect against cytokine-mediated cartilage resorption. This and other actions may underlie the potential benefits in treating OA. The exact relationships of these various actions, mostly determined in vitro, have not been specifically defined in vivo or ex vivo in relation to clinical efficacy. OUTCOMES HCQ and CQ have a good reputation for being effective and relatively safe treatments in SLE, mild-moderate RA and Sjøgren's syndrome. There is need for (a) more information on their mode of action in relation to the control of these diseases, (b) scope for developing formulations that have improved pharmacokinetic and therapeutic properties and safety, and (c) further exploring their use in drug combinations not only with other disease modifying agents but also with biologics.
Collapse
Affiliation(s)
- K D Rainsford
- Biomedical Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Ann L Parke
- Department of Rheumatology, St Francis Hospital and Medical Center, Hartford, CT, 06105, USA
| | | | - W F Kean
- Department of Medicine (Rheumatology), McMaster University Faculty of Health Sciences, Hamilton, ON, L8S 4K9, Canada.
- Department of Medicine (Rheumatology), McMaster University Faculty of Health Sciences, Suite #708, 1 Young Street, Hamilton, ON, L8N 1T8, Canada.
| |
Collapse
|
16
|
Pouwer RH, Deydier SM, Le PV, Schwartz BD, Franken NC, Davis RA, Coster MJ, Charman SA, Edstein MD, Skinner-Adams TS, Andrews KT, Jenkins ID, Quinn RJ. Total synthesis of thiaplakortone a: derivatives as metabolically stable leads for the treatment of malaria. ACS Med Chem Lett 2014; 5:178-82. [PMID: 24900794 DOI: 10.1021/ml400447v] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/27/2013] [Indexed: 01/26/2023] Open
Abstract
Thiaplakortone A (3a), an antimalarial natural product, was prepared by an operationally simple and scalable synthesis. In our efforts to deliver a lead compound with improved potency, metabolic stability, and selectivity, the synthesis was diverted to access a series of analogues. Compounds 3a-d showed nanomolar activity against the chloroquine-sensitive (3D7) Plasmodium falciparum line and were more active against the chloroquine- and mefloquine-resistant (Dd2) P. falciparum line. All compounds are "Rule-of-5" compliant, and we show that metabolic stability can be enhanced via modification at either the primary or pyrrole nitrogen. These promising results lay the foundation for the development of this structurally unprecedented natural product.
Collapse
Affiliation(s)
- Rebecca H. Pouwer
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Sophie M. Deydier
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Phuc Van Le
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Brett D. Schwartz
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Nicole C. Franken
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Rohan A. Davis
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Mark J. Coster
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Susan A. Charman
- Centre
for Drug Candidate Optimisation, Monash University, 381 Royal
Parade, Parkville, VIC 3052, Australia
| | | | - Tina S. Skinner-Adams
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Katherine T. Andrews
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Ian D. Jenkins
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Ronald J. Quinn
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
17
|
Hobbs CV, Tanaka TQ, Muratova O, Van Vliet J, Borkowsky W, Williamson KC, Duffy PE. HIV treatments have malaria gametocyte killing and transmission blocking activity. J Infect Dis 2013; 208:139-48. [PMID: 23539746 DOI: 10.1093/infdis/jit132] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Millions of individuals being treated for human immunodeficiency virus (HIV) live in malaria-endemic areas, but the effects of these treatments on malaria transmission are unknown. While drugs like HIV protease inhibitors (PIs) and trimethoprim-sulfamethoxazole (TMP-SMX) have known activity against parasites during liver or asexual blood stages, their effects on transmission stages require further study. METHODS The HIV PIs lopinavir and saquinavir, the nonnucleoside reverse-transcriptase inhibitor nevirapine, and the antibiotic TMP-SMX were assessed for activity against Plasmodium falciparum transmission stages. The alamarBlue assay was used to determine the effects of drugs on gametocyte viability, and exflagellation was assessed to determine the effects of drugs on gametocyte maturation. The effects of drug on transmission were assessed by calculating the mosquito oocyst count as a marker for infectivity, using standard membrane feeding assays. RESULTS Lopinavir and saquinavir have gametocytocidal and transmission blocking activities at or approaching clinically relevant treatment levels, while nevirapine does not. TMP-SMX is not gametocytocidal, but at prophylactic levels it blocks transmission. CONCLUSIONS Specific HIV treatments have gametocyte killing and transmission-blocking effects. Clinical studies are warranted to evaluate these findings and their potential impact on eradication efforts.
Collapse
Affiliation(s)
- Charlotte V Hobbs
- Laboratory of Malaria Vaccinology and Immunology, NIH/NIAID, 12735 Twinbrook Pkwy, Rockville, MD 20852, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Khan T, van Brummelen AC, Parkinson CJ, Hoppe HC. ATP and luciferase assays to determine the rate of drug action in in vitro cultures of Plasmodium falciparum. Malar J 2012; 11:369. [PMID: 23134617 PMCID: PMC3505462 DOI: 10.1186/1475-2875-11-369] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 10/30/2012] [Indexed: 11/10/2022] Open
Abstract
Background Knowledge of the rate of action of compounds against cultured malaria parasites is required to determine the optimal time-points for drug mode of action studies, as well as to predict likely in vivo parasite clearance rates in order to select optimal hit compounds for further development. In this study, changes in parasite ATP levels and transgenic luciferase reporter activity were explored as means to detect drug-induced stress in cultured parasites. Methods In vitro cultures of Plasmodium falciparum 3D7 wild-type or firefly luciferase-expressing parasites were incubated with a panel of six anti-malarial compounds for 10 hours and parasite ATP levels or luciferase activity determined at two-hour intervals using luminescence-based reagents. For comparative purposes, parasite morphology changes were evaluated by light microscopy, as well as the extent to which parasites recover after 48 hours from a six-hour drug treatment using a parasite lactate dehydrogenase assay. Results Changes in parasite ATP levels displayed three phenotypes: mild or no change (chloroquine, DFMO); 2–4 fold increase (mefloquine, artemisinin); severe depletion (ritonavir, gramicidin). The respective phenotypes and the rate at which they manifested correlated closely with the extent to which parasites recovered from a six-hour drug treatment (with the exception of chloroquine) and the appearance and severity of morphological changes observed by light microscopy. Luciferase activity decreased profoundly in parasites treated with mefloquine, artemisinin and ritonavir (34-67% decrease in 2 hours), while chloroquine and DFMO produced only mild changes over 10 hours. Gramicidin yielded intermediate decreases in luciferase activity. Conclusions ATP levels and luciferase activity respond rapidly to incubation with anti-malarial drugs and provide quantitative read-outs to detect the appearance and magnitude of drug-induced stress in cultured parasites. The correlation between the observed changes and irreversible parasite toxicity is not yet sufficiently clear to predict clinical clearance rates, but may be useful for ranking compounds against each other and standard drugs vis-à-vis rate of action and for determining early time-points for drug mode of action studies.
Collapse
Affiliation(s)
- Tasmiyah Khan
- CSIR Biosciences, PO Box 365, Pretoria 0001, South Africa
| | | | | | | |
Collapse
|
19
|
Alam A, Goyal M, Iqbal MS, Pal C, Dey S, Bindu S, Maity P, Bandyopadhyay U. Novel antimalarial drug targets: hope for new antimalarial drugs. Expert Rev Clin Pharmacol 2012; 2:469-89. [PMID: 22112223 DOI: 10.1586/ecp.09.28] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malaria is a major global threat, that results in more than 2 million deaths each year. The treatment of malaria is becoming extremely difficult due to the emergence of drug-resistant parasites, the absence of an effective vaccine, and the spread of insecticide-resistant vectors. Thus, malarial therapy needs new chemotherapeutic approaches leading to the search for new drug targets. Here, we discuss different approaches to identifying novel antimalarial drug targets. We have also given due attention to the existing validated targets with a view to develop novel, rationally designed lead molecules. Some of the important parasite proteins are claimed to be the targets; however, further in vitro or in vivo structure-function studies of such proteins are crucial to validate these proteins as suitable targets. The interactome analysis among apicoplast, mitochondrion and genomic DNA will also be useful in identifying vital pathways or proteins regulating critical pathways for parasite growth and survival, and could be attractive targets. Molecules responsible for parasite invasion to host erythrocytes and ion channels of infected erythrocytes, essential for intra-erythrocyte survival and stage progression of parasites are also becoming attractive targets. This review will discuss and highlight the current understanding regarding the potential antimalarial drug targets, which could be utilized to develop novel antimalarials.
Collapse
Affiliation(s)
- Athar Alam
- Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Synthesis and evaluation of hybrid drugs for a potential HIV/AIDS-malaria combination therapy. Bioorg Med Chem 2012; 20:5277-89. [PMID: 22858300 DOI: 10.1016/j.bmc.2012.06.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 06/13/2012] [Accepted: 06/21/2012] [Indexed: 02/06/2023]
Abstract
Malaria and HIV are among the most important global health problems of our time and together are responsible for approximately 3 million deaths annually. These two diseases overlap in many regions of the world including sub-Saharan Africa, Southeast Asia and South America, leading to a higher risk of co-infection. In this study, we generated and characterized hybrid molecules to target Plasmodium falciparum and HIV simultaneously for a potential HIV/malaria combination therapy. Hybrid molecules were synthesized by the covalent fusion of azidothymidine (AZT) with dihydroartemisinin (DHA), a tetraoxane or a 4-aminoquinoline derivative; and the small library was tested for antiviral and antimalarial activity. Our data suggests that compound 7 is the most potent molecule in vitro, with antiplasmodial activity comparable to that of DHA (IC(50)=26 nM, SI>3000), a moderate activity against HIV (IC(50)=2.9 μM; SI>35) and not toxic to HeLa cells at concentrations used in the assay (CC(50)>100 μM). Pharmacokinetics studies further revealed that compound 7 is metabolically unstable and is cleaved via O-dealkylation. These studies account for the lack of in vivo efficacy of compound 7 against the CQ-sensitive Plasmodium berghei N strain in mice, when administered orally at 20mg/kg.
Collapse
|
21
|
Antimalarial activity of the anticancer histone deacetylase inhibitor SB939. Antimicrob Agents Chemother 2012; 56:3849-56. [PMID: 22508312 DOI: 10.1128/aac.00030-12] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Histone deacetylase (HDAC) enzymes posttranslationally modify lysines on histone and nonhistone proteins and play crucial roles in epigenetic regulation and other important cellular processes. HDAC inhibitors (e.g., suberoylanilide hydroxamic acid [SAHA; also known as vorinostat]) are used clinically to treat some cancers and are under investigation for use against many other diseases. Development of new HDAC inhibitors for noncancer indications has the potential to be accelerated by piggybacking onto cancer studies, as several HDAC inhibitors have undergone or are undergoing clinical trials. One such compound, SB939, is a new orally active hydroxamate-based HDAC inhibitor with an improved pharmacokinetic profile compared to that of SAHA. In this study, the in vitro and in vivo antiplasmodial activities of SB939 were investigated. SB939 was found to be a potent inhibitor of the growth of Plasmodium falciparum asexual-stage parasites in vitro (50% inhibitory concentration [IC(50)], 100 to 200 nM), causing hyperacetylation of parasite histone and nonhistone proteins. In combination with the aspartic protease inhibitor lopinavir, SB939 displayed additive activity. SB939 also potently inhibited the in vitro growth of exoerythrocytic-stage Plasmodium parasites in liver cells (IC(50), ~150 nM), suggesting that inhibitor targeting to multiple malaria parasite life cycle stages may be possible. In an experimental in vivo murine model of cerebral malaria, orally administered SB939 significantly inhibited P. berghei ANKA parasite growth, preventing development of cerebral malaria-like symptoms. These results identify SB939 as a potent new antimalarial HDAC inhibitor and underscore the potential of investigating next-generation anticancer HDAC inhibitors as prospective new drug leads for treatment of malaria.
Collapse
|
22
|
Skinner-Adams TS, Butterworth AS, Porter KA, D'Amico R, Sawe F, Shaffer D, Siika A, Hosseinipour MC, Stringer E, Currier JS, Chipato T, Salata R, Lockman S, Eron JJ, Meshnick SR, McCarthy JS. The frequency of malaria is similar among women receiving either lopinavir/ritonavir or nevirapine-based antiretroviral treatment. PLoS One 2012; 7:e34399. [PMID: 22509297 PMCID: PMC3317955 DOI: 10.1371/journal.pone.0034399] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 02/27/2012] [Indexed: 11/19/2022] Open
Abstract
HIV protease inhibitors (PIs) show antimalarial activity in vitro and in animals. Whether this translates into a clinical benefit in HIV-infected patients residing in malaria-endemic regions is unknown. We studied the incidence of malaria, as defined by blood smear positivity or a positive Plasmodium falciparum histidine-rich protein 2 antigen test, among 444 HIV-infected women initiating antiretroviral treatment (ART) in the OCTANE trial (A5208; ClinicalTrials.gov: NCT00089505). Participants were randomized to treatment with PI-containing vs. PI-sparing ART, and were followed prospectively for ≥48 weeks; 73% also received cotrimoxazole prophylaxis. PI-containing treatment was not associated with protection against malaria in this study population.
Collapse
Affiliation(s)
- Tina S Skinner-Adams
- Infectious Diseases Division, Queensland Institute of Medical Research, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Saquinavir inhibits the malaria parasite's chloroquine resistance transporter. Antimicrob Agents Chemother 2012; 56:2283-9. [PMID: 22354298 DOI: 10.1128/aac.00166-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The antiretroviral protease inhibitors (APIs) ritonavir, saquinavir, and lopinavir, used to treat HIV infection, inhibit the growth of Plasmodium falciparum at clinically relevant concentrations. Moreover, it has been reported that these APIs potentiate the activity of chloroquine (CQ) against this parasite in vitro. The mechanism underlying this effect is not understood, but the degree of chemosensitization varies between the different APIs and, with the exception of ritonavir, appears to be dependent on the parasite exhibiting a CQ-resistant phenotype. Here we report a study of the role of the P. falciparum chloroquine resistance transporter (PfCRT) in the interaction between CQ and APIs, using transgenic parasites expressing different PfCRT alleles and using the Xenopus laevis oocyte system for the heterologous expression of PfCRT. Our data demonstrate that saquinavir behaves as a CQ resistance reverser and that this explains, at least in part, its ability to enhance the effects of CQ in CQ-resistant P. falciparum parasites.
Collapse
|
24
|
HIV-1 protease inhibitors and clinical malaria: a secondary analysis of the AIDS Clinical Trials Group A5208 study. Antimicrob Agents Chemother 2011; 56:995-1000. [PMID: 22123685 DOI: 10.1128/aac.05322-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV-1 protease inhibitors (PIs) have antimalarial activity in vitro and in murine models. The potential beneficial effect of HIV-1 PIs on malaria has not been studied in clinical settings. We used data from Adult AIDS Clinical Trials Group A5208 sites where malaria is endemic to compare the incidence of clinically diagnosed malaria among HIV-infected adult women randomized to either lopinavir/ritonavir (LPV/r)-based antiretroviral therapy (ART) or to nevirapine (NVP)-based ART. We calculated hazard ratios and 95% confidence intervals. We conducted a recurrent events analysis that included both first and second clinical malarial episodes and also conducted analyses to assess the sensitivity of results to outcome misclassification. Among the 445 women in this analysis, 137 (31%) received a clinical diagnosis of malaria at least once during follow-up. Of these 137, 72 (53%) were randomized to LPV/r-based ART. Assignment to the LPV/r treatment group (n = 226) was not consistent with a large decrease in the hazard of first clinical malarial episode (hazard ratio = 1.11 [0.79 to 1.56]). The results were similar in the recurrent events analysis. Sensitivity analyses indicated the results were robust to reasonable levels of outcome misclassification. In this study, the treatment with LPV/r compared to NVP had no apparent beneficial effect on the incidence of clinical malaria among HIV-infected adult women. Additional research concerning the effects of PI-based therapy on the incidence of malaria diagnosed by more specific criteria and among groups at a higher risk for severe disease is warranted.
Collapse
|
25
|
Hobbs C, Duffy P. Drugs for malaria: something old, something new, something borrowed. F1000 BIOLOGY REPORTS 2011; 3:24. [PMID: 22076126 PMCID: PMC3206709 DOI: 10.3410/b3-24] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Malaria was estimated to cause 800,000 deaths and 225 million cases worldwide in 2010. Worryingly, the first-line treatment currently relies on a single drug class called artemisinins, and there are signs that the parasite is becoming resistant to these drugs. The good news is that new technology has given us new approaches to drug discovery. New drugs generated this way are probably 10-15 years away from the clinic. Other antimalarials that may offer hope include those rehabilitated after not being used for some time, those that act as inhibitors of resistance mechanisms, those that limit infection while allowing protective immunity to develop, and those which are drugs borrowed from other disease treatments. All of these offer new hope of turning the tables on malaria. In parallel with the effort to develop vaccines that interrupt malaria transmission, drugs that target the parasite during transmission to the mosquito or during its pre-erythrocytic development in the liver, may allow us to terminate the parasite's spread.
Collapse
Affiliation(s)
- Charlotte Hobbs
- NIH/NIAID, Laboratory of Malaria Immunology and Vaccinology12735 Twinbrook Parkway, 3W19E, Rockville, MD 20852USA
| | - Patrick Duffy
- NIH/NIAID, Laboratory of Malaria Immunology and Vaccinology, Division of Intramural Research5640 Fishers Lane, Rm. 1111 Rockville, MD 20892USA
| |
Collapse
|
26
|
Flateau C, Le Loup G, Pialoux G. Consequences of HIV infection on malaria and therapeutic implications: a systematic review. THE LANCET. INFECTIOUS DISEASES 2011; 11:541-56. [PMID: 21700241 DOI: 10.1016/s1473-3099(11)70031-7] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite recent changes in the epidemiology of HIV infection and malaria and major improvements in their control, these diseases remain two of the most important infectious diseases and global health priorities. As they have overlapping distribution in tropical areas, particularly sub-Saharan Africa, any of their clinical, diagnostic, and therapeutic interactions might have important effects on patient care and public health policy. The biological basis of these interactions is well established. HIV infection induces cellular depletion and early abnormalities of CD4+ T cells, decreases CD8+ T-cell counts and function (cellular immunity), causes deterioration of specific antigen responses (humoral immunity), and leads to alteration of innate immunity through impairment of cytolytic activity and cytokine production by natural killer cells. Therefore, HIV infection affects the immune response to malaria, particularly premunition in adolescents and adults, and pregnancy-specific immunity, leading to different patterns of disease in HIV-infected patients compared with HIV-uninfected patients. In this systematic review, we collate data on the effects of HIV on malaria and discuss their therapeutic consequences. HIV infection is associated with increased prevalence and severity of clinical malaria and impaired response to antimalarial treatment, depending on age, immunodepression, and previous immunity to malaria. HIV also affects pregnancy-specific immunity to malaria and response to intermittent preventive treatment. Co-trimoxazole (trimethoprim-sulfamethoxazole) prophylaxis and antiretroviral treatment reduce occurrence of clinical malaria; however, these therapies interact with antimalarial drugs, and new therapeutic guidelines are needed for concomitant use.
Collapse
Affiliation(s)
- Clara Flateau
- Service des Maladies Infectieuses et Tropicales, Hôpital Tenon, AP-HP, University Pierre et Marie Curie, Paris, France
| | | | | |
Collapse
|
27
|
Synergy of the antiretroviral protease inhibitor indinavir and chloroquine against malaria parasites in vitro and in vivo. Parasitol Res 2011; 109:1519-24. [PMID: 21537980 DOI: 10.1007/s00436-011-2427-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 04/08/2011] [Indexed: 10/18/2022]
Abstract
Many malaria-endemic areas are also associated with high rates of human immunodeficiency virus (HIV) infection. An understanding of the chemotherapeutic interactions that occur during malaria and HIV co-infections is important. Our previous studies have demonstrated that some antiretroviral protease inhibitors are effective in inhibiting Plasmodium falciparum growth in vitro. Currently, studies examining the interactions between antiretroviral protease inhibitors and antimalarial drugs are being conducted, but the data are limited. In this study, we examined the synergistic interactions between the antiretroviral protease inhibitor indinavir and chloroquine (CQ) in chloroquine-resistant and chloroquine-sensitive malaria parasites in vitro and in vivo. In vitro, by using modified fixed-ratio isobologram method, fractional inhibitory concentrations index (FICI) was calculated to indicate the interaction between the two drugs. The results demonstrated that indinavir interacted synergistically with chloroquine against both chloroquine-sensitive P. falciparum clone 3D7 (mean FICI 0.784) and multidrug-resistant P. falciparum clone Dd2 (mean FICI 0.599). In vivo drug interactions were measured using a 4-day suppressive test in a rodent malaria model infected with Plasmodium chabaudi. We observed that indinavir enhanced the antimalarial activity of chloroquine against both the chloroquine-sensitive line P. chabaudi ASS and the chloroquine-resistant line P. chabaudi ASCQ. More importantly, chloroquine had a 100% clearance of asexual parasites when used in combination with indinavir at an appropriate dose ratio (10 mg/kg CQ + 1.8 g/kg indinavir) where there was no obvious toxicity. We conclude from this study that the combination of indinavir and chloroquine may become a novel antimalarial drug regimen.
Collapse
|
28
|
Grimberg BT, Mehlotra RK. Expanding the Antimalarial Drug Arsenal-Now, But How? Pharmaceuticals (Basel) 2011; 4:681-712. [PMID: 21625331 PMCID: PMC3102560 DOI: 10.3390/ph4050681] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 04/09/2011] [Accepted: 04/19/2011] [Indexed: 01/24/2023] Open
Abstract
The number of available and effective antimalarial drugs is quickly dwindling. This is mainly because a number of drug resistance-associated mutations in malaria parasite genes, such as crt, mdr1, dhfr/dhps, and others, have led to widespread resistance to all known classes of antimalarial compounds. Unfortunately, malaria parasites have started to exhibit some level of resistance in Southeast Asia even to the most recently introduced class of drugs, artemisinins. While there is much need, the antimalarial drug development pipeline remains woefully thin, with little chemical diversity, and there is currently no alternative to the precious artemisinins. It is difficult to predict where the next generation of antimalarial drugs will come from; however, there are six major approaches: (i) re-optimizing the use of existing antimalarials by either replacement/rotation or combination approach; (ii) repurposing drugs that are currently used to treat other infections or diseases; (iii) chemically modifying existing antimalarial compounds; (iv) exploring natural sources; (v) large-scale screening of diverse chemical libraries; and (vi) through parasite genome-based ("targeted") discoveries. When any newly discovered effective antimalarial treatment is used by the populus, we must maintain constant vigilance for both parasite-specific and human-related factors that are likely to hamper its success. This article is neither comprehensive nor conclusive. Our purpose is to provide an overview of antimalarial drug resistance, associated parasite genetic factors (1. Introduction; 2. Emergence of artemisinin resistance in P. falciparum), and the antimalarial drug development pipeline (3. Overview of the global pipeline of antimalarial drugs), and highlight some examples of the aforementioned approaches to future antimalarial treatment. These approaches can be categorized into "short term" (4. Feasible options for now) and "long term" (5. Next generation of antimalarial treatment-Approaches and candidates). However, these two categories are interrelated, and the approaches in both should be implemented in parallel with focus on developing a successful, long-lasting antimalarial chemotherapy.
Collapse
Affiliation(s)
- Brian T. Grimberg
- Center for Global Health and Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; E-Mails: (B.T.G.); (R.K.M.); Tel.: +1-216-368-6328 or +1-216-368-6172, Fax: +1-216-368-4825
| | - Rajeev K. Mehlotra
- Center for Global Health and Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; E-Mails: (B.T.G.); (R.K.M.); Tel.: +1-216-368-6328 or +1-216-368-6172, Fax: +1-216-368-4825
| |
Collapse
|
29
|
Antimalarial effects of human immunodeficiency virus protease inhibitors in rhesus macaques. Antimicrob Agents Chemother 2011; 55:3039-42. [PMID: 21486958 DOI: 10.1128/aac.00085-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The antimalarial activity of the human immunodeficiency virus protease inhibitors indinavir and saquinavir was evaluated in rhesus macaques for the first time. Indinavir effectively suppressed the growth of Plasmodium cynomolgi and Plasmodium knowlesi in vivo after a 7- or 3-day treatment, respectively, with clinically relevant doses, whereas saquinavir showed only weak activity against P. cynomolgi.
Collapse
|
30
|
Alfonso Y, Monzote L. HIV Protease Inhibitors: Effect on the Opportunistic Protozoan Parasites. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2011; 5:40-50. [PMID: 21629510 PMCID: PMC3103880 DOI: 10.2174/1874104501105010040] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 05/22/2010] [Accepted: 06/28/2010] [Indexed: 11/22/2022]
Abstract
The impact of highly active antiretroviral therapy (HAART) in the natural history of AIDS disease has been allowed to prolong the survival of people with HIV infection, particularly whose with increased HIV viral load. Additionally, the antiretroviral therapy could exert a certain degree of protection against parasitic diseases. A number of studies have been evidenced a decrease in the incidence of opportunistic parasitic infections in the era of HAART. Although these changes have been attributed to the restoration of cell-mediated immunity, induced by either non-nucleoside reverse transcriptase inhibitors or HIV protease inhibitors, in combination with at least two nucleoside reverse transcriptase inhibitors included in HAART, there are evidence that the control of these parasitic infections in HIV-positive persons under HAART, is also induced by the inhibition of the proteases of the parasites. This review focuses on the principal available data related with therapeutic HIV-protease inhibitors and their in vitro and in vivo effects on the opportunistic protozoan parasites.
Collapse
Affiliation(s)
- Yenisey Alfonso
- Parasitology Department, Institute of Tropical Medicine “Pedro Kourí”, Cuba
| | | |
Collapse
|
31
|
White RE, Powell DJ, Berry C. HIV proteinase inhibitors target the Ddi1-like protein of Leishmania parasites. FASEB J 2011; 25:1729-36. [PMID: 21266539 PMCID: PMC3739880 DOI: 10.1096/fj.10-178947] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
HIV proteinase inhibitors reduce the levels of Leishmania parasites in vivo and in vitro, but their biochemical target is unknown. We have identified an ortholog of the yeast Ddi1 protein as the only member of the aspartic proteinase family in Leishmania parasites, and in this study we investigate this protein as a potential target for the drugs. To date, no enzyme assay has been developed for the Ddi1 proteins, but Saccharomyces cerevisiae lacking the DDI1 gene secrete high levels of protein into the medium. We developed an assay in which these knockout yeast were functionally complemented to low secretion by introduction of genes encoding Ddi1 orthologs from Leishmania major or humans. Plasmid alone controls gave no complementation. Treatment of the Ddi1 transformants with HIV proteinase inhibitors showed differential effects dependent on the origin of the Ddi1. Dose responses allowed calculation of IC50 values; e.g., for nelfinavir, of 3.4 μM (human Ddi1) and 0.44 μM (Leishmania Ddi1). IC50 values with Leishmania constructs mirror the potency of inhibitors against parasites. Our results show that Ddi1 proteins are targets of HIV proteinase inhibitors and indicates the Leishmania Ddi1 as the likely target for these drugs and a potential target for antiparasitic therapy.—White, R. E., Powell, D. J., Berry, C. HIV proteinase inhibitors target the Ddi1-Like protein of Leishmania parasites.
Collapse
Affiliation(s)
- Rhian E White
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | | | |
Collapse
|
32
|
Karp CL, Mahanty S. Approach to the Patient with HIV and Coinfecting Tropical Infectious Diseases. TROPICAL INFECTIOUS DISEASES: PRINCIPLES, PATHOGENS AND PRACTICE 2011. [PMCID: PMC7150329 DOI: 10.1016/b978-0-7020-3935-5.00139-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Guantai EM, Ncokazi K, Egan TJ, Gut J, Rosenthal PJ, Smith PJ, Chibale K. Design, synthesis and in vitro antimalarial evaluation of triazole-linked chalcone and dienone hybrid compounds. Bioorg Med Chem 2010; 18:8243-56. [PMID: 21044845 DOI: 10.1016/j.bmc.2010.10.009] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 09/27/2010] [Accepted: 10/05/2010] [Indexed: 11/28/2022]
Abstract
A targeted series of chalcone and dienone hybrid compounds containing aminoquinoline and nucleoside templates was synthesized and evaluated for in vitro antimalarial activity. The Cu(I)-catalyzed cycloaddition of azides and terminal alkynes was applied as the hybridization strategy. Several chalcone-chloroquinoline hybrid compounds were found to be notably active, with compound 8b the most active, exhibiting submicromolar IC(50) values against the D10, Dd2 and W2 strains of Plasmodium falciparum.
Collapse
Affiliation(s)
- Eric M Guantai
- Department of Chemistry, University of Cape Town, South Africa
| | | | | | | | | | | | | |
Collapse
|
34
|
Gardiner DL, Skinner-Adams TS, Brown CL, Andrews KT, Stack CM, McCarthy JS, Dalton JP, Trenholme KR. Plasmodium falciparum: new molecular targets with potential for antimalarial drug development. Expert Rev Anti Infect Ther 2010; 7:1087-98. [PMID: 19883329 DOI: 10.1586/eri.09.93] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malaria remains one of the world's most devastating infectious diseases. Drug resistance to all classes of antimalarial agents has now been observed, highlighting the need for new agents that act against novel parasite targets. The complete sequencing of the Plasmodium falciparum genome has allowed the identification of new molecular targets within the parasite that may be amenable to chemotherapeutic intervention. In this review, we investigate four possible targets for the future development of new classes of antimalarial agents. These targets include histone deacetylase, the aspartic proteases or plasmepsins, aminopeptidases and the purine salvage enzyme hypoxanthine-xanthine-guanine phosphoribosyltransferase.
Collapse
Affiliation(s)
- Donald L Gardiner
- Malaria Biology Laboratory, Queensland Institute of Medical Research, 300 Herston Road, Herston, QLD 4006, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Mishra LC, Bhattacharya A, Sharma M, Bhasin VK. HIV protease inhibitors, indinavir or nelfinavir, augment antimalarial action of artemisinin in vitro. Am J Trop Med Hyg 2010; 82:148-50. [PMID: 20065012 DOI: 10.4269/ajtmh.2010.09-0427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Most malaria endemic regions are co-infested with HIV infection. Treatment of one may affect outcome of the other in co-infected individuals. HIV protease inhibitors, indinavir or nelfinavir, are important antiretroviral drugs and artemisinin is central to malaria treatment. We show these protease inhibitors augment the antimalarial activity of artemisinin against P. falciparum in vitro.
Collapse
|
36
|
Antimalarial asexual stage-specific and gametocytocidal activities of HIV protease inhibitors. Antimicrob Agents Chemother 2009; 54:1334-7. [PMID: 20028821 DOI: 10.1128/aac.01512-09] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The stage-specific antimalarial activities of a panel of antiretroviral protease inhibitors (PIs), including two nonpeptidic PIs (tipranavir and darunavir), were tested in vitro against Plasmodium falciparum. While darunavir demonstrated limited antimalarial activity (effective concentration [EC(50)], >50 microM), tipranavir was active at clinically relevant concentrations (EC(50), 12 to 21 microM). Saquinavir, lopinavir, and tipranavir preferentially inhibited the growth of mature asexual-stage parasites (24 h postinvasion). While all of the PIs tested inhibited gametocytogenesis, tipranavir was the only one to exhibit gametocytocidal activity.
Collapse
|
37
|
In vitro interactions between antiretroviral protease inhibitors and artemisinin endoperoxides against Plasmodium falciparum. Int J Antimicrob Agents 2009; 35:191-3. [PMID: 19896339 DOI: 10.1016/j.ijantimicag.2009.09.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 11/21/2022]
Abstract
Antiretroviral protease inhibitors (APIs), which are effective at controlling the effects of human immunodeficiency virus (HIV) in patients, have also proven efficacious in inhibiting Plasmodium falciparum growth in vitro. Use of artemisinin-based combination therapies is being encouraged to reduce malaria mortality in areas of P. falciparum resistance to conventional antimalarial drugs. The aim of this study was to investigate drug interactions between HIV protease inhibitors and artemisinin drugs against malaria. In vitro cultures of P. falciparum provide a screen system for identifying and evaluating drug combinations. The derived isobolograms provide clear evidence of antagonistic interactions between artemisinin endoperoxides and several different APIs.
Collapse
|
38
|
He Z, Chen L, You J, Qin L, Chen X. Antiretroviral protease inhibitors potentiate chloroquine antimalarial activity in malaria parasites by regulating intracellular glutathione metabolism. Exp Parasitol 2009; 123:122-7. [DOI: 10.1016/j.exppara.2009.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 06/05/2009] [Accepted: 06/10/2009] [Indexed: 11/26/2022]
|
39
|
The Impact of HIV and Malaria Coinfection: What Is Known and Suggested Venues for Further Study. Interdiscip Perspect Infect Dis 2009; 2009:617954. [PMID: 19680452 PMCID: PMC2723755 DOI: 10.1155/2009/617954] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 06/01/2009] [Indexed: 01/27/2023] Open
Abstract
HIV and malaria have similar global distributions. Annually, 500 million are infected and 1 million die because of malaria. 33 million have HIV and 2 million die from it each year. Minor effects of one infection on the disease course or outcome for the other would significantly impact public health because of the sheer number of people at risk for coinfection. While early population-based studies showed no difference in outcomes between HIV-positive and HIV-negative individuals with malaria, more recent work suggests that those with HIV have more frequent episodes of symptomatic malaria and that malaria increases HIV plasma viral load and decreases CD4+ T cells. HIV and malaria each interact with the host's immune system, resulting in a complex activation of immune cells, and subsequent dysregulated production of cytokines and antibodies. Further investigation of these interactions is needed to better define effects of coinfection.
Collapse
|
40
|
Hobbs CV, Voza T, Coppi A, Kirmse B, Marsh K, Borkowsky W, Sinnis P. HIV protease inhibitors inhibit the development of preerythrocytic-stage plasmodium parasites. J Infect Dis 2009; 199:134-41. [PMID: 19032102 DOI: 10.1086/594369] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Recent studies have demonstrated that human immunodeficiency virus (HIV) protease inhibitors (PIs) exert inhibitory effects on erythrocytic stages of the human-malaria parasite Plasmodium falciparum in vitro and on erythrocytic stages of the rodent-malaria parasite Plasmodium chabaudi in vivo. Although it remains unclear how HIV PIs inhibit the parasite, the effect seen on parasite development in the erythrocytic stages is potent. The effect on preerythrocytic stages has not yet been investigated. Using the rodent parasite Plasmodium berghei, we screened a panel of HIV PIs in vitro for effects on the preerythrocytic stages. Our data indicated that the HIV PIs lopinavir and saquinavir affect preerythrocytic-stage parasite development in vitro. We then evaluated the effect of HIV PIs on preerythrocytic stages in vivo using the rodent parasite Plasmodium yoelii. We found that lopinavir/ritonavir had a dose-dependent effect on liver-stage parasite development. Given that sub-Saharan Africa is where the HIV/AIDS pandemic intersects with malaria, these results merit analysis in clinical settings.
Collapse
Affiliation(s)
- Charlotte V Hobbs
- NYU School of Medicine, Dept. of Pediatrics, Div. of Infectious Disease, 550 First Ave., 8W New Bellevue, NY, NY 10010, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Lek-Uthai U, Suwanarusk R, Ruengweerayut R, Skinner-Adams TS, Nosten F, Gardiner DL, Boonma P, Piera KA, Andrews KT, Machunter B, McCarthy JS, Anstey NM, Price RN, Russell B. Stronger activity of human immunodeficiency virus type 1 protease inhibitors against clinical isolates of Plasmodium vivax than against those of P. falciparum. Antimicrob Agents Chemother 2008; 52:2435-41. [PMID: 18443130 PMCID: PMC2443880 DOI: 10.1128/aac.00169-08] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 03/26/2008] [Accepted: 04/22/2008] [Indexed: 11/20/2022] Open
Abstract
Recent studies using laboratory clones have demonstrated that several antiretroviral protease inhibitors (PIs) inhibit the growth of Plasmodium falciparum at concentrations that may be of clinical significance, especially during human immunodeficiency virus type 1 (HIV-1) and malaria coinfection. Using clinical isolates, we now demonstrate the in vitro effectiveness of two HIV-1 aspartic PIs, saquinavir (SQV) and ritonavir (RTV), against P. vivax (n = 30) and P. falciparum (n = 20) from populations subjected to high levels of mefloquine and artesunate pressure on the Thailand-Myanmar border. The median 50% inhibitory concentration values of P. vivax to RTV and SQV were 2,233 nM (range, 732 to 7,738 nM) and 4,230 nM (range, 1,326 to 8,452 nM), respectively, both within the therapeutic concentration range commonly found for patients treated with these PIs. RTV was fourfold more effective at inhibiting P. vivax than it was at inhibiting P. falciparum, compared to a twofold difference in SQV sensitivity. An increased P. falciparum mdr1 copy number was present in 33% (3/9) of isolates and that of P. vivax mdr1 was present in 9% of isolates (2/22), but neither was associated with PI sensitivity. The inter-Plasmodium sp. variations in PI sensitivity indicate key differences between P. vivax and P. falciparum. PI-containing antiretroviral regimens may demonstrate prophylactic activity against both vivax and falciparum malaria in HIV-infected patients who reside in areas where multidrug-resistant P. vivax or P. falciparum is found.
Collapse
Affiliation(s)
- U Lek-Uthai
- Department of Parasitology, Faculty of Public Health, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Skinner-Adams TS, McCarthy JS, Gardiner DL, Andrews KT. HIV and malaria co-infection: interactions and consequences of chemotherapy. Trends Parasitol 2008; 24:264-71. [PMID: 18456554 DOI: 10.1016/j.pt.2008.03.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 02/12/2008] [Accepted: 03/10/2008] [Indexed: 10/22/2022]
Abstract
The global epidemiology of HIV/AIDS and malaria overlap because a significant number of HIV-infected individuals live in regions with different levels of malaria transmission. Although the consequences of co-infection with HIV and malaria parasites are not fully understood, available evidence suggests that the infections act synergistically and together result in worse outcomes. The importance of understanding chemotherapeutic interactions during malaria and HIV co-infection is now being recognized. We know that some antimalarial drugs have weak antiretroviral effects; however, recent studies have also demonstrated that certain antiretroviral agents can inhibit malaria-parasite growth. Here, we discuss recent findings on the impact of HIV/AIDS and malaria co-infection and the possible roles of chemotherapy in improving the treatment of these diseases.
Collapse
|
43
|
Synergy of human immunodeficiency virus protease inhibitors with chloroquine against Plasmodium falciparum in vitro and Plasmodium chabaudi in vivo. Antimicrob Agents Chemother 2008; 52:2653-6. [PMID: 18443126 DOI: 10.1128/aac.01329-07] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The synergy of the activities between chloroquine and various human immunodeficiency virus protease inhibitors was investigated in chloroquine-resistant and -sensitive malaria parasites. In both in vitro and in vivo assay systems, ritonavir was found to be the most potent in potentiating the antimalarial action of chloroquine.
Collapse
|
44
|
Bonilla JA, Bonilla TD, Yowell CA, Fujioka H, Dame JB. Critical roles for the digestive vacuole plasmepsins ofPlasmodium falciparumin vacuolar function. Mol Microbiol 2007; 65:64-75. [PMID: 17581121 DOI: 10.1111/j.1365-2958.2007.05768.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Knockout mutants of Plasmodium falciparum lacking pfpm1, pfpm2 and pfhap (triple-PM KO), and mutants lacking all four digestive vacuole (DV) plasmepsins (pfpm4, pfpm1, pfpm2 and pfhap; quadruple-PM KO), were prepared by double cross-over integration effecting chromosomal deletions of up to 14.6 kb. The triple-PM KO was similar to the parental line (3D7) in growth rate, morphology and sensitivity to proteinase inhibitors. The quadruple-PM KO showed a significantly slower rate of growth in standard medium, which manifested as delayed schizont maturation accompanied by reduced formation of haemozoin. In amino acid-limited medium, the reduction in growth rate of the quadruple-PM KO was pronounced. The sensitivity of both the triple- and quadruple-PM KOs to six different HIV aspartic proteinase inhibitors was comparable to that of 3D7, thus establishing that the DV plasmepsins were not the primary targets of the antimalarial activity of these clinically important compounds. Electron microscopic analysis revealed the presence of multilamellar bodies resembling ceroid in the DV of the quadruple-PM KO, and intermediates of the autophagic pathway accumulated as determined by Western blot analysis. Thus, the DV plasmepsins, although not essential, contribute significantly to the fitness of the parasite and are required for efficient degradation of endosomal vesicles delivered to the DV.
Collapse
Affiliation(s)
- J Alfredo Bonilla
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32611, USA
| | | | | | | | | |
Collapse
|
45
|
Andrews KT, Gatton ML, Skinner-Adams TS, McCarthy JS, Gardiner DL. HIV-malaria interactions: don't forget the drugs. Science 2007; 315:1791; author reply 1791. [PMID: 17395812 DOI: 10.1126/science.315.5820.1791a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|