1
|
Markus MB. Putative Contribution of 8-Aminoquinolines to Preventing Recrudescence of Malaria. Trop Med Infect Dis 2023; 8:278. [PMID: 37235326 PMCID: PMC10223033 DOI: 10.3390/tropicalmed8050278] [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: 04/12/2023] [Revised: 05/07/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Enhanced therapeutic efficacy achieved in treating Plasmodium vivax malaria with an 8-aminoquinoline (8-AQ) drug such as primaquine (PQ) together with a partner drug such as chloroquine (CQ) is usually explained as CQ inhibiting asexual parasites in the bloodstream and PQ acting against liver stages. However, PQ's contribution, if any, to inactivating non-circulating, extra-hepatic asexual forms, which make up the bulk of the parasite biomass in chronic P. vivax infections, remains unclear. In this opinion article, I suggest that, considering its newly described mode of action, PQ might be doing something of which we are currently unaware.
Collapse
Affiliation(s)
- Miles B. Markus
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg 2193, South Africa;
- School of Animal, Plant and Environmental Sciences, Faculty of Science, University of Witwatersrand, Johannesburg 2001, South Africa
| |
Collapse
|
2
|
Bakadlag R, Limniatis G, Georges G, Georges E. The anti-estrogen receptor drug, tamoxifen, is selectively Lethal to P-glycoprotein-expressing Multidrug resistant tumor cells. BMC Cancer 2023; 23:24. [PMID: 36609245 PMCID: PMC9824978 DOI: 10.1186/s12885-022-10474-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND P-glycoprotein (P-gp), a member of the ATP Binding Cassette B1 subfamily (ABCB1), confers resistance to clinically relevant anticancer drugs and targeted chemotherapeutics. However, paradoxically P-glycoprotein overexpressing drug resistant cells are "collaterally sensitive" to non-toxic drugs that stimulate its ATPase activity. METHODS Cell viability assays were used to determine the effect of low concentrations of tamoxifen on the proliferation of multidrug resistant cells (CHORC5 and MDA-Doxo400), expressing P-gp, their parental cell lines (AuxB1 and MDA-MB-231) or P-gp-CRISPR knockout clones of AuxB1 and CHORC5 cells. Western blot analysis was used to estimate P-gp expression in different cell lines. Apoptosis of tamoxifen-induced cell death was estimated by flow cytometry using Annexin-V-FITC stained cells. Oxidative stress of tamoxifen treated cells was determined by measuring levels of reactive oxygen species and reduced thiols using cell-permeant 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) and 5,5-dithio-bis-(2-nitrobenzoic acid) DTNB, respectively. RESULTS In this report, we show that P-gp-expressing drug resistant cells (CHORC5 and MDA-Doxo400) are collaterally sensitive to the anti-estrogen tamoxifen or its metabolite (4-hydroxy-tamoxifen). Moreover, P-gp-knockout clones of CHORC5 cells display complete reversal of collateral sensitivity to tamoxifen. Drug resistant cells exposed to low concentrations of tamoxifen show significant rise in reactive oxygen species, drop of reduced cellular thiols and increased apoptosis. Consistent with the latter, CHORC5 cells expressing high levels of human Bcl-2 (CHORC5-Bcl-2) show significant resistance to tamoxifen. In addition, the presence of the antioxidant N-acetylcysteine or P-gp ATPase inhibitor, PSC-833, reverse the collateral sensitivity of resistant cells to tamoxifen. By contrast, the presence of rotenone (specific inhibitor of mitochondria complex I) synergizes with tamoxifen. CONCLUSION This study demonstrates the use of tamoxifen as collateral sensitivity drug that can preferentially target multidrug resistant cells expressing P-gp at clinically achievable concentrations. Given the widespread use of tamoxifen in the treatment of estrogen receptor-positive breast cancers, this property of tamoxifen may have clinical applications in treatment of P-gp-positive drug resistant breast tumors.
Collapse
Affiliation(s)
- Rowa Bakadlag
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, Ste. Anne de Bellevue, Québec, H9X-3V9 Canada
| | - Georgia Limniatis
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, Ste. Anne de Bellevue, Québec, H9X-3V9 Canada
| | - Gabriel Georges
- grid.421142.00000 0000 8521 1798Department of Cardiac Surgery, Quebec Heart & Lung Institute, Université Laval, Québec, Canada
| | - Elias Georges
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, Ste. Anne de Bellevue, Québec, H9X-3V9 Canada
| |
Collapse
|
3
|
New In Vitro Interaction-Parasite Reduction Ratio Assay for Early Derisk in Clinical Development of Antimalarial Combinations. Antimicrob Agents Chemother 2022; 66:e0055622. [PMID: 36197116 PMCID: PMC9664866 DOI: 10.1128/aac.00556-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The development and spread of drug-resistant phenotypes substantially threaten malaria control efforts. Combination therapies have the potential to minimize the risk of resistance development but require intensive preclinical studies to determine optimal combination and dosing regimens. To support the selection of new combinations, we developed a novel in vitro-in silico combination approach to help identify the pharmacodynamic interactions of the two antimalarial drugs in a combination which can be plugged into a pharmacokinetic/pharmacodynamic model built with human monotherapy parasitological data to predict the parasitological endpoints of the combination. This makes it possible to optimally select drug combinations and doses for the clinical development of antimalarials. With this assay, we successfully predicted the endpoints of two phase 2 clinical trials in patients with the artefenomel-piperaquine and artefenomel-ferroquine drug combinations. In addition, the predictive performance of our novel in vitro model was equivalent to that of the humanized mouse model outcome. Last, our more informative in vitro combination assay provided additional insights into the pharmacodynamic drug interactions compared to the in vivo systems, e.g., a concentration-dependent change in the maximum killing effect (Emax) and the concentration producing 50% of the killing maximum effect (EC50) of piperaquine or artefenomel or a directional reduction of the EC50 of ferroquine by artefenomel and a directional reduction of Emax of ferroquine by artefenomel. Overall, this novel in vitro-in silico-based technology will significantly improve and streamline the economic development of new drug combinations for malaria and potentially also in other therapeutic areas.
Collapse
|
4
|
Komatsuya K, Sakura T, Shiomi K, Ōmura S, Hikosaka K, Nozaki T, Kita K, Inaoka DK. Siccanin Is a Dual-Target Inhibitor of Plasmodium falciparum Mitochondrial Complex II and Complex III. Pharmaceuticals (Basel) 2022; 15:ph15070903. [PMID: 35890202 PMCID: PMC9319939 DOI: 10.3390/ph15070903] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/05/2023] Open
Abstract
Plasmodium falciparum contains several mitochondrial electron transport chain (ETC) dehydrogenases shuttling electrons from the respective substrates to the ubiquinone pool, from which electrons are consecutively transferred to complex III, complex IV, and finally to the molecular oxygen. The antimalarial drug atovaquone inhibits complex III and validates this parasite’s ETC as an attractive target for chemotherapy. Among the ETC dehydrogenases from P. falciparum, dihydroorotate dehydrogenase, an essential enzyme used in de novo pyrimidine biosynthesis, and complex III are the two enzymes that have been characterized and validated as drug targets in the blood-stage parasite, while complex II has been shown to be essential for parasite survival in the mosquito stage; therefore, these enzymes and complex II are considered candidate drug targets for blocking parasite transmission. In this study, we identified siccanin as the first (to our knowledge) nanomolar inhibitor of the P. falciparum complex II. Moreover, we demonstrated that siccanin also inhibits complex III in the low-micromolar range. Siccanin did not inhibit the corresponding complexes from mammalian mitochondria even at high concentrations. Siccanin inhibited the growth of P. falciparum with IC50 of 8.4 μM. However, the growth inhibition of the P. falciparum blood stage did not correlate with ETC inhibition, as demonstrated by lack of resistance to siccanin in the yDHODH-3D7 (EC50 = 10.26 μM) and Dd2-ELQ300 strains (EC50 = 18.70 μM), suggesting a third mechanism of action that is unrelated to mitochondrial ETC inhibition. Hence, siccanin has at least a dual mechanism of action, being the first potent and selective inhibitor of P. falciparum complexes II and III over mammalian enzymes and so is a potential candidate for the development of a new class of antimalarial drugs.
Collapse
Affiliation(s)
- Keisuke Komatsuya
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; (K.K.); (T.N.)
- Laboratory of Biomembrane, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takaya Sakura
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan;
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
| | - Kazuro Shiomi
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo 108-8641, Japan;
| | - Satoshi Ōmura
- Ōmura Satoshi Memorial Institute, Kitasato University, Minato-ku, Tokyo 108-8641, Japan;
| | - Kenji Hikosaka
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; (K.K.); (T.N.)
| | - Kiyoshi Kita
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; (K.K.); (T.N.)
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
- Department of Host-Defense Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Correspondence: (K.K.); (D.K.I.); Tel.: +81-95-819-7575 (K.K.); +81-95-819-7230 (D.K.I.)
| | - Daniel Ken Inaoka
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; (K.K.); (T.N.)
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan;
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
- Correspondence: (K.K.); (D.K.I.); Tel.: +81-95-819-7575 (K.K.); +81-95-819-7230 (D.K.I.)
| |
Collapse
|
5
|
A Hybrid of Amodiaquine and Primaquine Linked by Gold(I) Is a Multistage Antimalarial Agent Targeting Heme Detoxification and Thiol Redox Homeostasis. Pharmaceutics 2022; 14:pharmaceutics14061251. [PMID: 35745823 PMCID: PMC9229949 DOI: 10.3390/pharmaceutics14061251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Hybrid-based drugs linked through a transition metal constitute an emerging concept for Plasmodium intervention. To advance the drug design concept and enhance the therapeutic potential of this class of drugs, we developed a novel hybrid composed of quinolinic ligands amodiaquine (AQ) and primaquine (PQ) linked by gold(I), named [AuAQPQ]PF6. This compound demonstrated potent and efficacious antiplasmodial activity against multiple stages of the Plasmodium life cycle. The source of this activity was thoroughly investigated by comparing parasite susceptibility to the hybrid's components, the annotation of structure-activity relationships and studies of the mechanism of action. The activity of [AuAQPQ]PF6 for the parasite's asexual blood stages was influenced by the presence of AQ, while its activity against gametocytes and pre-erythrocytic parasites was influenced by both quinolinic components. Moreover, the coordination of ligands to gold(I) was found to be essential for the enhancement of potency, as suggested by the observation that a combination of quinolinic ligands does not reproduce the antimalarial potency and efficacy as observed for the metallic hybrid. Our results indicate that this gold(I) hybrid compound presents a dual mechanism of action by inhibiting the beta-hematin formation and enzymatic activity of thioredoxin reductases. Overall, our findings support the potential of transition metals as a dual chemical linker and an antiplasmodial payload for the development of hybrid-based drugs.
Collapse
|
6
|
Verma L, Vekilov PG, Palmer JC. Solvent Structure and Dynamics near the Surfaces of β-Hematin Crystals. J Phys Chem B 2021; 125:11264-11274. [PMID: 34609878 DOI: 10.1021/acs.jpcb.1c06589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hematin crystallization, which is an essential component of the physiology of malaria parasites and the most successful target for antimalarial drugs, proceeds in mixed organic-aqueous solvents both in vivo and in vitro. Here we employ molecular dynamics simulations to examine the structuring and dynamics of a water-normal octanol mixture (a solvent that mimics the environment hosting hematin crystallization in vivo) in the vicinity of the typical faces in the habit of a hematin crystal. The simulations reveal that the properties of the solvent in the layer adjacent to the crystal are strongly impacted by the distinct chemical and topological features presented by each crystal face. The solvent organizes into at least three distinct layers. We also show that structuring of the solvent near the different faces of β-hematin strongly impacts the interfacial dynamics. The relaxation time of n-octanol molecules is longest in the contact layers and correlates with the degree of structural ordering at the respective face. We show that the macroscopically homogeneous water-octanol solution holds clusters of water and n-octanol connected by hydrogen bonds that entrap the majority of the water but are mostly smaller than 30 water molecules. Near the crystal surface the clusters anchor on hematin carboxyl groups. These results provide a direct example that solvent structuring is not restricted to aqueous and other hydrogen-bonded solutions. Our findings illuminate two fundamental features of the mechanisms of hematin crystallization: the elongated shapes of natural and synthetic hematin crystals and the stabilization of charged groups of hematin and antimalarials by encasing in water clusters. In addition, these findings suggest that hematin crystallization may be controlled by additives that disrupt or reinforce solvent structuring.
Collapse
Affiliation(s)
- Laksmanji Verma
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| | - Peter G Vekilov
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States.,Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Jeremy C Palmer
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
7
|
Artemisinin-Based Drugs Target the Plasmodium falciparum Heme Detoxification Pathway. Antimicrob Agents Chemother 2021; 65:AAC.02137-20. [PMID: 33495226 DOI: 10.1128/aac.02137-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/16/2021] [Indexed: 12/18/2022] Open
Abstract
Artemisinin (ART)-based antimalarial drugs are believed to exert lethal effects on malarial parasites by alkylating a variety of intracellular molecular targets. Recent work with live parasites has shown that one of the alkylated targets is free heme within the parasite digestive vacuole, which is liberated upon hemoglobin catabolism by the intraerythrocytic parasite, and that reduced levels of heme alkylation occur in artemisinin-resistant parasites. One implication of heme alkylation is that these drugs may inhibit parasite detoxification of free heme via inhibition of heme-to-hemozoin crystallization; however, previous reports that have investigated this hypothesis present conflicting data. By controlling reducing conditions and, hence, the availability of ferrous versus ferric forms of free heme, we modify a previously reported hemozoin inhibition assay to quantify the ability of ART-based drugs to target the heme detoxification pathway under reduced versus oxidizing conditions. Contrary to some previous reports, we find that artemisinins are potent inhibitors of hemozoin crystallization, with effective half-maximal concentrations approximately an order of magnitude lower than those for most quinoline-based antimalarial drugs. We also examine hemozoin and in vitro parasite growth inhibition for drug pairs found in the most commonly used ART-based combination therapies (ACTs). All ACTs examined inhibit hemozoin crystallization in an additive fashion, and all but one inhibit parasite growth in an additive fashion.
Collapse
|
8
|
Nutmakul T, Pattanapanyasat K, Soonthornchareonnon N, Shiomi K, Mori M, Prathanturarug S. Speed of action and stage specificity of Bencha-loga-wichian, a Thai traditional antipyretic formulation, against Plasmodium falciparum and the chloroquine-potentiating activity of its active compounds, tiliacorinine and yanangcorinine. JOURNAL OF ETHNOPHARMACOLOGY 2020; 258:112909. [PMID: 32360802 DOI: 10.1016/j.jep.2020.112909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/02/2020] [Accepted: 04/21/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bencha-loga-wichian (BLW), a Thai traditional antipyretic formulation, has been reported to have promising antiplasmodial activity, and it was previously revealed that tiliacorinine and yanangcorinine, isolated from Tiliacora triandra, were the active compounds. However, the mechanisms of action of BLW have not been investigated. In addition, these active compounds are bisbenzylisoquinoline alkaloids, many compounds of which have been reported to potentiate the efficacy of chloroquine. AIMS OF THE STUDY To investigate the antiplasmodial mechanisms of action of BLW and evaluate the effects of chloroquine combined with tiliacorinine or yanangcorinine. MATERIALS AND METHODS Chloroquine-resistant Plasmodium falciparum (PfW2) strains at the ring, trophozoite, and schizont stages were exposed to the extracts or compounds for 2, 4, 6, 8, 10, 12, 24 or 48 h. The percentages of parasitemia were determined by flow cytometry, and their morphologies were examined by Giemsa-stained smear to evaluate the speed of action and stage specificity. For the drug combination assay, a modified fixed-ratio isobologram method was used. RESULTS The antiplasmodial activity of BLW possessed a slow onset of action and was the most effective against ring-stage parasites. After 48 h of extracts or compounds exposure, most of the treated parasites, at all stages, turned to the pyknotic form and could not recover even after extracts or compounds removal. The results suggested that these extracts and compounds could kill the parasites or possess parasiticidal effects. In addition, the combination of chloroquine with tiliacorinine or yanangcorinine demonstrated a synergistic effect, indicating that these compounds could potentiate chloroquine efficacy against chloroquine-resistant parasites. CONCLUSION The antiplasmodial mechanisms of action of BLW appeared to differ from that of chloroquine and other current antimalarial drugs. In addition, tiliacorinine and yanangcorinine, the active compounds of BLW, could potentiate the efficacy of chloroquine. Accordingly, BLW was shown to be a good candidate for development as a new antimalarial and useful for drug combination therapy.
Collapse
Affiliation(s)
- Thanutchaporn Nutmakul
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Road, Bangkok, 10400, Thailand.
| | - Kovit Pattanapanyasat
- Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Noppamas Soonthornchareonnon
- Department of Pharmacognosy, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Road, Bangkok, 10400, Thailand.
| | - Kazuro Shiomi
- Laboratory of Biological Functions, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| | - Mihoko Mori
- Laboratory of Biological Functions, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| | - Sompop Prathanturarug
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Road, Bangkok, 10400, Thailand.
| |
Collapse
|
9
|
Antagonistic cooperativity between crystal growth modifiers. Nature 2020; 577:497-501. [DOI: 10.1038/s41586-019-1918-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022]
|
10
|
Huang RY, Pei L, Liu Q, Chen S, Dou H, Shu G, Yuan ZX, Lin J, Peng G, Zhang W, Fu H. Isobologram Analysis: A Comprehensive Review of Methodology and Current Research. Front Pharmacol 2019; 10:1222. [PMID: 31736746 PMCID: PMC6830115 DOI: 10.3389/fphar.2019.01222] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/23/2019] [Indexed: 01/12/2023] Open
Abstract
Drug combination is a common method for clinical disease treatment. Whether the combination of drugs is reasonable often affects the result of the disease treatment. Many methods have been used to evaluate interaction between drugs to date. Isobologram analysis has been mathematically proven and widely used to evaluate drug interactions. In this paper, the principle of isobologram analysis and its application in drug interaction evaluation are summarized. The applications of the similar cotoxicity coefficient and fractional inhibitory concentration index in the evaluation of drug interaction are also reviewed. This work is expected to evaluate the effect of formulations scientifically and provide scientific judgment standards for the development of formulations and clinical drug compatibility.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hualin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agriculture University, Chengdu, China
| |
Collapse
|
11
|
Viability Screen of LOPAC 1280 Reveals Tyrosine Kinase Inhibitor Tyrphostin A9 as a Novel Partner Drug for Artesunate Combinations To Target the Plasmodium falciparum Ring Stage. Antimicrob Agents Chemother 2019; 63:AAC.02389-18. [PMID: 30718250 DOI: 10.1128/aac.02389-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/21/2019] [Indexed: 01/25/2023] Open
Abstract
The emergence of artemisinin-resistant Plasmodium falciparum poses a major threat to current frontline artemisinin combination therapies. Artemisinin resistance is widely associated with mutations in the P. falciparum Kelch13 (PfKelch13) propeller region, leading to delayed parasite clearance and increased survival of early-ring-stage parasites. There is therefore a need to discover novel drugs that are effective against artemisinin-resistant P. falciparum In view of this, our study aimed to identify compounds from the Library of Pharmacologically Active Compounds1280 (LOPAC1280) that could increase the efficacy of artesunate and be used as a potential partner drug for treatment against artemisinin-resistant falciparum malaria. By using a modified ring-stage survival assay, we performed a high-throughput screening of the activities of the 1,280 compounds from the LOPAC library in combination with artesunate against the P. falciparum IPC 5202 field isolate harboring the R539T mutation in the PfKelch13 propeller region. The potencies of the hits against both the IPC 5202 and CamWT_C580Y field isolates were determined through dose-dependent isobologram analyses; CamWT_C580Y has the more prevalent C580Y mutation characteristic of strains with artemisinin resistance. We identified tyrphostin A9 to have synergistic and additive activity against both parasite strains when dosed in combination with artesunate. These findings provide promising novel artesunate combinations that can target the P. falciparum artemisinin-resistant ring stage and insights that may aid in obtaining a better understanding of the mechanism involved in artemisinin resistance.
Collapse
|
12
|
Kirkman LA, Zhan W, Visone J, Dziedziech A, Singh PK, Fan H, Tong X, Bruzual I, Hara R, Kawasaki M, Imaeda T, Okamoto R, Sato K, Michino M, Alvaro EF, Guiang LF, Sanz L, Mota DJ, Govindasamy K, Wang R, Ling Y, Tumwebaze PK, Sukenick G, Shi L, Vendome J, Bhanot P, Rosenthal PJ, Aso K, Foley MA, Cooper RA, Kafsack B, Doggett JS, Nathan CF, Lin G. Antimalarial proteasome inhibitor reveals collateral sensitivity from intersubunit interactions and fitness cost of resistance. Proc Natl Acad Sci U S A 2018; 115:E6863-E6870. [PMID: 29967165 PMCID: PMC6055138 DOI: 10.1073/pnas.1806109115] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We describe noncovalent, reversible asparagine ethylenediamine (AsnEDA) inhibitors of the Plasmodium falciparum proteasome (Pf20S) β5 subunit that spare all active subunits of human constitutive and immuno-proteasomes. The compounds are active against erythrocytic, sexual, and liver-stage parasites, against parasites resistant to current antimalarials, and against P. falciparum strains from patients in Africa. The β5 inhibitors synergize with a β2 inhibitor in vitro and in mice and with artemisinin. P. falciparum selected for resistance to an AsnEDA β5 inhibitor surprisingly harbored a point mutation in the noncatalytic β6 subunit. The β6 mutant was resistant to the species-selective Pf20S β5 inhibitor but remained sensitive to the species-nonselective β5 inhibitors bortezomib and carfilzomib. Moreover, resistance to the Pf20S β5 inhibitor was accompanied by increased sensitivity to a Pf20S β2 inhibitor. Finally, the β5 inhibitor-resistant mutant had a fitness cost that was exacerbated by irradiation. Thus, used in combination, multistage-active inhibitors of the Pf20S β5 and β2 subunits afford synergistic antimalarial activity with a potential to delay the emergence of resistance to artemisinins and each other.
Collapse
Affiliation(s)
- Laura A Kirkman
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, NY 10065;
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065
| | - Wenhu Zhan
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065
| | - Joseph Visone
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, NY 10065
| | - Alexis Dziedziech
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, NY 10065
| | - Pradeep K Singh
- Chemical Core Facility, Department of Biochemistry, Weill Cornell Medicine, NY 10065
| | - Hao Fan
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065
| | - Xinran Tong
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065
| | - Igor Bruzual
- Department of Research and Development, Portland Veterans Affairs Medical Center, Oregon Health and Science University, Portland, OR 97239
| | - Ryoma Hara
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Masanori Kawasaki
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Toshihiro Imaeda
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Rei Okamoto
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Kenjiro Sato
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Elena Fernandez Alvaro
- Diseases of the Developing World (DDW), Tres Cantos Medicine Development Campus, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Liselle F Guiang
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901
| | - Laura Sanz
- Diseases of the Developing World (DDW), Tres Cantos Medicine Development Campus, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Daniel J Mota
- Department of Medicine, University of California, San Francisco, CA 94143
| | - Kavitha Govindasamy
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 11201
| | - Rong Wang
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Yan Ling
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065
| | | | - George Sukenick
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Lei Shi
- Department of Biophysics, Weill Cornell Medicine, NY 10065
| | | | - Purnima Bhanot
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 11201
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA 94143
| | - Kazuyoshi Aso
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Michael A Foley
- Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065
| | - Roland A Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901
| | - Bjorn Kafsack
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065
| | - J Stone Doggett
- Department of Research and Development, Portland Veterans Affairs Medical Center, Oregon Health and Science University, Portland, OR 97239
| | - Carl F Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065;
| | - Gang Lin
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY 10065;
| |
Collapse
|
13
|
Aguiar ACC, Panciera M, Simão dos Santos EF, Singh MK, Garcia ML, de Souza GE, Nakabashi M, Costa JL, Garcia CRS, Oliva G, Correia CRD, Guido RVC. Discovery of Marinoquinolines as Potent and Fast-Acting Plasmodium falciparum Inhibitors with in Vivo Activity. J Med Chem 2018; 61:5547-5568. [DOI: 10.1021/acs.jmedchem.8b00143] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Anna Caroline Campos Aguiar
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Michele Panciera
- Institute of Chemistry, State University of Campinas, Josue de Castro St., Campinas, SP 13083-970, Brazil
| | | | - Maneesh Kumar Singh
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Cidade Universitária, São Paulo, SP 05508-900, Brazil
- Department of Physiology, University of Sao Paulo, Rua do Matão 101, Travessa 14, São Paulo, SP 05508-090, Brazil
| | - Mariana Lopes Garcia
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Guilherme Eduardo de Souza
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Myna Nakabashi
- Department of Physiology, University of Sao Paulo, Rua do Matão 101, Travessa 14, São Paulo, SP 05508-090, Brazil
| | - José Luiz Costa
- Faculty of Pharmaceutical Sciences, State University of Campinas, Rua Oswaldo Cruz, 2° Andar, Bloco F3, Cidade Universitária, Campinas, SP 13083-859, Brazil
| | - Célia R. S. Garcia
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Cidade Universitária, São Paulo, SP 05508-900, Brazil
| | - Glaucius Oliva
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | | | - Rafael Victorio Carvalho Guido
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| |
Collapse
|
14
|
Chan C, Martin P, Liptrott NJ, Siccardi M, Almond L, Owen A. Incompatibility of chemical protein synthesis inhibitors with accurate measurement of extended protein degradation rates. Pharmacol Res Perspect 2018; 5. [PMID: 28971619 PMCID: PMC5625163 DOI: 10.1002/prp2.359] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/15/2017] [Indexed: 12/29/2022] Open
Abstract
Protein synthesis inhibitors are commonly used for measuring protein degradation rates, but may cause cytotoxicity via direct or indirect mechanisms. This study aimed to identify concentrations providing optimal inhibition in the absence of overt cytotoxicity. Actinomycin D, cycloheximide, emetine, and puromycin were assessed individually, and in two-, three-, and four-drug combinations for protein synthesis inhibition (IC50 ) and cytotoxicity (CC50 ) over 72 h. Experiments were conducted in HepG2 cells and primary rat hepatocytes (PRH). IC50 for actinomycin D, cycloheximide, emetine, and puromycin were 39 ± 7.4, 6600 ± 2500, 2200 ± 1400, and 1600 ± 1200 nmol/L; with corresponding CC50 values of 6.2 ± 7.3, 570 ± 510, 81 ± 9, and 1300 ± 64 nmol/L, respectively, in HepG2 cells. The IC50 were 1.7 ± 1.8, 290 ± 90, 620 ± 920, and 2000 ± 2000 nmol/L, with corresponding CC50 values of 0.98 ± 1.8, 680 ± 1300, 180 ± 700, and 1600 ± 1000 (SD) nmol/L, respectively, in PRH. CC50 were also lower than the IC50 for all drug combinations in HepG2 cells. These data indicate that using pharmacological interference is inappropriate for measuring protein degradation over a protracted period, because inhibitory effects cannot be extricated from cytotoxicity.
Collapse
Affiliation(s)
- Christina Chan
- Department of Molecular and Clinical Pharmacology, The University of Liverpool, 70 Pembroke Place, Block H (first floor), Liverpool, L69 3GF, United Kingdom
| | - Philip Martin
- Department of Molecular and Clinical Pharmacology, The University of Liverpool, 70 Pembroke Place, Block H (first floor), Liverpool, L69 3GF, United Kingdom
| | - Neill J Liptrott
- Department of Molecular and Clinical Pharmacology, The University of Liverpool, 70 Pembroke Place, Block H (first floor), Liverpool, L69 3GF, United Kingdom
| | - Marco Siccardi
- Department of Molecular and Clinical Pharmacology, The University of Liverpool, 70 Pembroke Place, Block H (first floor), Liverpool, L69 3GF, United Kingdom
| | - Lisa Almond
- Simcyp (a Certara company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, United Kingdom
| | - Andrew Owen
- Department of Molecular and Clinical Pharmacology, The University of Liverpool, 70 Pembroke Place, Block H (first floor), Liverpool, L69 3GF, United Kingdom
| |
Collapse
|
15
|
Parra LLL, Bertonha AF, Severo IRM, Aguiar ACC, de Souza GE, Oliva G, Guido RVC, Grazzia N, Costa TR, Miguel DC, Gadelha FR, Ferreira AG, Hajdu E, Romo D, Berlinck RGS. Isolation, Derivative Synthesis, and Structure-Activity Relationships of Antiparasitic Bromopyrrole Alkaloids from the Marine Sponge Tedania brasiliensis. JOURNAL OF NATURAL PRODUCTS 2018; 81:188-202. [PMID: 29297684 PMCID: PMC5989537 DOI: 10.1021/acs.jnatprod.7b00876] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The isolation and identification of a series of new pseudoceratidine (1) derivatives from the sponge Tedania brasiliensis enabled the evaluation of their antiparasitic activity against Plasmodium falciparum, Leishmania (Leishmania) amazonensis, Leishmania (Leishmania) infantum, and Trypanosoma cruzi, the causative agents of malaria, cutaneous leishmaniasis, visceral leishmaniasis, and Chagas disease, respectively. The new 3-debromopseudoceratidine (4), 20-debromopseudoceratidine (5), 4-bromopseudoceratidine (6), 19-bromopseudoceratidine (7), and 4,19-dibromopseudoceratidine (8) are reported. New tedamides A-D (9-12), with an unprecedented 4-bromo-4-methoxy-5-oxo-4,5-dihydro-1H-pyrrole-2-carboxamide moiety, are also described. Compounds 4 and 5, 6 and 7, 9 and 10, and 11 and 12 have been isolated as pairs of inseparable structural isomers differing in their sites of bromination or oxidation. Tedamides 9+10 and 11+12 were obtained as optically active pairs, indicating an enzymatic formation rather than an artifactual origin. N12-Acetylpseudoceratidine (2) and N12-formylpseudoceratidine (3) were obtained by derivatization of pseudoceratidine (1). The antiparasitic activity of pseudoceratidine (1) led us to synthesize 23 derivatives (16, 17, 20, 21, 23, 25, 27-29, 31, 33, 35, 38, 39, 42, 43, 46, 47, 50, and 51) with variations in the polyamine chain and aromatic moiety in sufficient amounts for biological evaluation in antiparasitic assays. The measured antimalarial activity of pseudoceratidine (1) and derivatives 4, 5, 16, 23, 25, 31, and 50 provided an initial SAR evaluation of these compounds as potential leads for antiparasitics against Leishmania amastigotes and against P. falciparum. The results obtained indicate that pseudoceratidine represents a promising scaffold for the development of new antimalarial drugs.
Collapse
Affiliation(s)
- Lizbeth L. L. Parra
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, CEP 13560-970, São Carlos, SP, Brazil
| | - Ariane F. Bertonha
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, CEP 13560-970, São Carlos, SP, Brazil
- Department of Chemistry & Biochemistry, Baylor University, Waco, TX 76706, USA
| | - Ivan R. M. Severo
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, CEP 13560-970, São Carlos, SP, Brazil
| | - Anna C. C. Aguiar
- Instituto de Física de São Carlos, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, SP, 13563-120, Brazil
| | - Guilherme E. de Souza
- Instituto de Física de São Carlos, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, SP, 13563-120, Brazil
| | - Glaucius Oliva
- Instituto de Física de São Carlos, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, SP, 13563-120, Brazil
| | - Rafael V. C. Guido
- Instituto de Física de São Carlos, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, SP, 13563-120, Brazil
| | - Nathalia Grazzia
- Departamento de Biologia Animal e Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, CEP 13083-862, Campinas, SP, Brazil
| | - Tábata R. Costa
- Departamento de Biologia Animal e Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, CEP 13083-862, Campinas, SP, Brazil
| | - Danilo C. Miguel
- Departamento de Biologia Animal e Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, CEP 13083-862, Campinas, SP, Brazil
| | - Fernanda R. Gadelha
- Departamento de Biologia Animal e Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, CEP 13083-862, Campinas, SP, Brazil
| | - Antonio G. Ferreira
- Departamento de Química, Universidade Federal de São Carlos, Rod. Washington Luiz, km 235 - SP-310, CEP 13565-905, São Carlos, SP, Brazil
| | - Eduardo Hajdu
- Museu Nacional, Universidade Federal do Rio de Janeiro, Quinta da Boa Vista, s/n, CEP 20940-040, Rio de Janeiro, RJ, Brazil
| | - Daniel Romo
- Department of Chemistry & Biochemistry, Baylor University, Waco, TX 76706, USA
| | - Roberto G. S. Berlinck
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, CEP 13560-970, São Carlos, SP, Brazil
| |
Collapse
|
16
|
Abugri DA, Witola WH, Jaynes JM. In vitro antagonistic and indifferent activity of combination of 3-deoxyanthocyanidins against Toxoplasma gondii. Parasitol Res 2017; 116:3387-3400. [PMID: 29086004 DOI: 10.1007/s00436-017-5661-1] [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] [Received: 03/21/2017] [Accepted: 10/18/2017] [Indexed: 10/18/2022]
Abstract
Toxoplasma gondii is a ubiquitous intracellular zoonotic parasite estimated to affect about 30-90% of the world's human population. The most affected are immunocompromised individuals such as HIV-AIDS and cancer patients, organ and tissue transplant recipients, and congenitally infected children. No effective and safe drugs and vaccines are available against all forms of the parasite. We report here the antagonistic and indifferent activity of the combination of five different formulations of pure synthetic 3-deoxyanthocyaninidin (3-DA) chloride compounds against T. gondii tachyzoites and the synergistic and additive interaction against a human foreskin fibroblast (HFF) cell line in vitro using fluorescence microscopy, trypan blue assay, and fractional inhibitory concentration index. The individual and the combined pure 3-DA compounds were observed to have effective inhibition against T. gondii parasites with less cytotoxic effect in a ratio of 1:1. The IC50 values for parasite inhibition ranged from 1.88 μg/mL (1.51-2.32 μg/mL) for luteolinindin plus 7-methoxyapigeninindin (LU/7-MAP) and 2.23 μg/mL (1.66-2.97 μg/mL) for apigeninindin plus 7-methoxyapigeninindin (AP/7-MAP) combinations at 95% confidence interval (CI) after 48 h of culture. We found LU/7-MAP to be antagonistic and AP/7-MAP to be indifferent in interaction against T. gondii growth. Both individual and combination 3-DA compounds not only depicted very strong inhibitory activity against T. gondii, but also had synergistic and additive cytotoxic effects against HFF cells. These synthetic 3-DAs have potential as antiparasitic agents for the treatment of human toxoplasmosis.
Collapse
Affiliation(s)
- Daniel A Abugri
- Department of Chemistry, College of Arts and Sciences, Laboratory of Ethnomedicine, Parasitology and Drug Discovery, Tuskegee University, Tuskegee, USA. .,Department of Biology, College of Arts and Sciences, Tuskegee University, Tuskegee, AL, USA.
| | - William H Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Jesse M Jaynes
- Department of Agricultural and Environmental Sciences, College of Agriculture, Environment and Nutrition Sciences, Tuskegee University, Tuskegee, AL, USA
| |
Collapse
|
17
|
Kemirembe K, Cabrera M, Cui L. Interactions between tafenoquine and artemisinin-combination therapy partner drug in asexual and sexual stage Plasmodium falciparum. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2017; 7:131-137. [PMID: 28319724 PMCID: PMC5358947 DOI: 10.1016/j.ijpddr.2017.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/04/2017] [Accepted: 03/08/2017] [Indexed: 12/18/2022]
Abstract
The 8-aminoquinoline tafenoquine (TFQ), a primaquine derivative, is currently in late-stage clinical development for the radical cure of P. vivax. Here drug interactions between TFQ and chloroquine and six artemisinin-combination therapy (ACT) partner drugs in P. falciparum asexual stages and gametocytes were investigated. TFQ was mostly synergistic with the ACT-partner drugs in asexual parasites regardless of genetic backgrounds. However, at fixed ratios of 1:3, 1:1 and 3:1, TFQ only interacted synergistically with naphthoquine, pyronaridine and piperaquine in gametocytes. This study indicated that TFQ and ACT-partner drugs will likely have increased potency against asexual stages of the malaria parasites, whereas some drugs may interfere with each other against the P. falciparum gametocytes.
Collapse
Affiliation(s)
- Karen Kemirembe
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Mynthia Cabrera
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Liwang Cui
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
18
|
Matthews H, Deakin J, Rajab M, Idris-Usman M, Nirmalan NJ. Investigating antimalarial drug interactions of emetine dihydrochloride hydrate using CalcuSyn-based interactivity calculations. PLoS One 2017; 12:e0173303. [PMID: 28257497 PMCID: PMC5336292 DOI: 10.1371/journal.pone.0173303] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/17/2017] [Indexed: 11/18/2022] Open
Abstract
The widespread introduction of artemisinin-based combination therapy has contributed to recent reductions in malaria mortality. Combination therapies have a range of advantages, including synergism, toxicity reduction, and delaying the onset of resistance acquisition. Unfortunately, antimalarial combination therapy is limited by the depleting repertoire of effective drugs with distinct target pathways. To fast-track antimalarial drug discovery, we have previously employed drug-repositioning to identify the anti-amoebic drug, emetine dihydrochloride hydrate, as a potential candidate for repositioned use against malaria. Despite its 1000-fold increase in in vitro antimalarial potency (ED50 47 nM) compared with its anti-amoebic potency (ED50 26–32 uM), practical use of the compound has been limited by dose-dependent toxicity (emesis and cardiotoxicity). Identification of a synergistic partner drug would present an opportunity for dose-reduction, thus increasing the therapeutic window. The lack of reliable and standardised methodology to enable the in vitro definition of synergistic potential for antimalarials is a major drawback. Here we use isobologram and combination-index data generated by CalcuSyn software analyses (Biosoft v2.1) to define drug interactivity in an objective, automated manner. The method, based on the median effect principle proposed by Chou and Talalay, was initially validated for antimalarial application using the known synergistic combination (atovaquone-proguanil). The combination was used to further understand the relationship between SYBR Green viability and cytocidal versus cytostatic effects of drugs at higher levels of inhibition. We report here the use of the optimised Chou Talalay method to define synergistic antimalarial drug interactivity between emetine dihydrochloride hydrate and atovaquone. The novel findings present a potential route to harness the nanomolar antimalarial efficacy of this affordable natural product.
Collapse
Affiliation(s)
- Holly Matthews
- Environment and Life sciences, University of Salford, Greater Manchester, United Kingdom
| | - Jon Deakin
- Environment and Life sciences, University of Salford, Greater Manchester, United Kingdom
| | - May Rajab
- Environment and Life sciences, University of Salford, Greater Manchester, United Kingdom
| | - Maryam Idris-Usman
- Environment and Life sciences, University of Salford, Greater Manchester, United Kingdom
| | - Niroshini J. Nirmalan
- Environment and Life sciences, University of Salford, Greater Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
19
|
Ferraz R, Noronha J, Murtinheira F, Nogueira F, Machado M, Prudêncio M, Parapini S, D'Alessandro S, Teixeira C, Gomes A, Prudêncio C, Gomes P. Primaquine-based ionic liquids as a novel class of antimalarial hits. RSC Adv 2016. [DOI: 10.1039/c6ra10759a] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ionic liquids derived from active pharmaceutical ingredients may open new perspectives towards low-cost rescuing of classical antimalarial drugs.
Collapse
|
20
|
In Vitro Activities of Primaquine-Schizonticide Combinations on Asexual Blood Stages and Gametocytes of Plasmodium falciparum. Antimicrob Agents Chemother 2015; 59:7650-6. [PMID: 26416869 DOI: 10.1128/aac.01948-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/23/2015] [Indexed: 12/28/2022] Open
Abstract
Currently, the World Health Organization recommends addition of a 0.25-mg base/kg single dose of primaquine (PQ) to artemisinin combination therapies (ACTs) for Plasmodium falciparum malaria as a gametocytocidal agent for reducing transmission. Here, we investigated the potential interactions of PQ with the long-lasting components of the ACT drugs for eliminating the asexual blood stages and gametocytes of in vitro-cultured P. falciparum strains. Using the SYBR green I assay for asexual parasites and a flow cytometry-based assay for gametocytes, we determined the interactions of PQ with the schizonticides chloroquine, mefloquine, piperaquine, lumefantrine, and naphthoquine. With the sums of fractional inhibitory concentrations and isobolograms, we were able to determine mostly synergistic interactions for the various PQ and schizonticide combinations on the blood stages of P. falciparum laboratory strains. The synergism in inhibiting asexual stages and gametocytes was highly evident with PQ-naphthoquine, whereas synergism was moderate for the PQ-piperaquine, PQ-chloroquine, and PQ-mefloquine combinations. We have detected potentially antagonistic interactions between PQ and lumefantrine under certain drug combination ratios, suggesting that precautions might be needed when PQ is added as the gametocytocide to the artemether-lumefantrine ACT (Coartem).
Collapse
|
21
|
Mott BT, Eastman RT, Guha R, Sherlach KS, Siriwardana A, Shinn P, McKnight C, Michael S, Lacerda-Queiroz N, Patel PR, Khine P, Sun H, Kasbekar M, Aghdam N, Fontaine SD, Liu D, Mierzwa T, Mathews-Griner LA, Ferrer M, Renslo AR, Inglese J, Yuan J, Roepe PD, Su XZ, Thomas CJ. High-throughput matrix screening identifies synergistic and antagonistic antimalarial drug combinations. Sci Rep 2015; 5:13891. [PMID: 26403635 PMCID: PMC4585899 DOI: 10.1038/srep13891] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/07/2015] [Indexed: 01/22/2023] Open
Abstract
Drug resistance in Plasmodium parasites is a constant threat. Novel therapeutics, especially new drug combinations, must be identified at a faster rate. In response to the urgent need for new antimalarial drug combinations we screened a large collection of approved and investigational drugs, tested 13,910 drug pairs, and identified many promising antimalarial drug combinations. The activity of known antimalarial drug regimens was confirmed and a myriad of new classes of positively interacting drug pairings were discovered. Network and clustering analyses reinforced established mechanistic relationships for known drug combinations and identified several novel mechanistic hypotheses. From eleven screens comprising >4,600 combinations per parasite strain (including duplicates) we further investigated interactions between approved antimalarials, calcium homeostasis modulators, and inhibitors of phosphatidylinositide 3-kinases (PI3K) and the mammalian target of rapamycin (mTOR). These studies highlight important targets and pathways and provide promising leads for clinically actionable antimalarial therapy.
Collapse
Affiliation(s)
- Bryan T. Mott
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Richard T. Eastman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Katy S. Sherlach
- Department of Chemistry, Georgetown University, 37th and O St., NW, Washington, DC
| | - Amila Siriwardana
- Department of Chemistry, Georgetown University, 37th and O St., NW, Washington, DC
| | - Paul Shinn
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Crystal McKnight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Sam Michael
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Norinne Lacerda-Queiroz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Paresma R. Patel
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Pwint Khine
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Hongmao Sun
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Monica Kasbekar
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Nima Aghdam
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
- Department of Chemistry, Georgetown University, 37th and O St., NW, Washington, DC
| | - Shaun D. Fontaine
- Department of Pharmaceutical Chemistry, Small Molecule Discovery Center, University of California, San Francisco, CA
| | - Dongbo Liu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Tim Mierzwa
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Lesley A. Mathews-Griner
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, Small Molecule Discovery Center, University of California, San Francisco, CA
| | - James Inglese
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Jing Yuan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Paul D. Roepe
- Department of Chemistry, Georgetown University, 37th and O St., NW, Washington, DC
- Department of Biochemistry, Cellular and Molecular Biology and Center for Infectious Diseases, Georgetown University, 37th and O St., NW, Washington, DC
| | - Xin-zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD
| |
Collapse
|
22
|
Yan C, Wei H, Minjuan Z, Yan X, Jingyue Y, Wenchao L, Sheng H. The mTOR inhibitor rapamycin synergizes with a fatty acid synthase inhibitor to induce cytotoxicity in ER/HER2-positive breast cancer cells. PLoS One 2014; 9:e97697. [PMID: 24866893 PMCID: PMC4035285 DOI: 10.1371/journal.pone.0097697] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/23/2014] [Indexed: 12/20/2022] Open
Abstract
Patients with ER/HER2-positive breast cancer have a poor prognosis and are less responsive to selective estrogen receptor modulators; this is presumably due to the crosstalk between ER and HER2. Fatty acid synthase (FASN) is essential for the survival and maintenance of the malignant phenotype of breast cancer cells. An intimate relationship exists between FASN, ER and HER2. We hypothesized that FASN may be the downstream effector underlying ER/HER2 crosstalk through the PI3K/AKT/mTOR pathway in ER/HER2-positive breast cancer. The present study implicated the PI3K/AKT/mTOR pathway in the regulation of FASN expression in ER/HER2-positive breast cancer cells and demonstrated that rapamycin, an mTOR inhibitor, inhibited FASN expression. Cerulenin, a FASN inhibitor, synergized with rapamycin to induce apoptosis and inhibit cell migration and tumorigenesis in ER/HER2-positive breast cancer cells. Our findings suggest that inhibiting the mTOR-FASN axis is a promising new strategy for treating ER/HER2-positive breast cancer.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cerulenin/pharmacology
- Drug Synergism
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Fatty Acid Synthase, Type I/antagonists & inhibitors
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Female
- Fluorescent Antibody Technique, Indirect
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice
- Mice, Nude
- Protein Kinase Inhibitors/pharmacology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sirolimus/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chen Yan
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Huang Wei
- Department of Cardiology, Xi'jing hospital, Fourth military medical university, Xi'an, PR China
| | - Zheng Minjuan
- Department of Ultrasound, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Xue Yan
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yang Jingyue
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Liu Wenchao
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
- * E-mail: (LW); (HS)
| | - Han Sheng
- State Key Laboratory of Military Stomatology, Department of Information Center, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
- * E-mail: (LW); (HS)
| |
Collapse
|
23
|
Roepe PD. To kill or not to kill, that is the question: cytocidal antimalarial drug resistance. Trends Parasitol 2014; 30:130-5. [PMID: 24530127 DOI: 10.1016/j.pt.2014.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 02/03/2023]
Abstract
Elucidating mechanisms of antimalarial drug resistance accelerates development of improved diagnostics and the design of new, effective malaria therapy. Recently, several studies have emphasized that chloroquine (CQ) resistance (CQR) can be quantified in two very distinct ways, depending on whether sensitivity to the growth inhibitory effects or parasite-kill effects of the drug are being measured. It is now clear that these cytostatic and cytocidal CQR phenotypes are not equivalent, and recent genetic, cell biological, and biophysical evidence suggests how the molecular mechanisms may overlap. These conclusions have important implications for elucidating other drug resistance phenomena and emphasize new concepts that are essential for the development of new drug therapy.
Collapse
Affiliation(s)
- Paul D Roepe
- Department of Chemistry, Georgetown University, 37th and O Streets NW, Washington DC 20057, USA; Department of Biochemistry and Cellular and Molecular Biology, Georgetown University, 37th and O Streets NW, Washington DC 20057, USA.
| |
Collapse
|