1
|
Rout S, Mahapatra RK. Plasmodium falciparum: Multidrug resistance. Chem Biol Drug Des 2019; 93:737-759. [DOI: 10.1111/cbdd.13484] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 01/05/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Subhashree Rout
- School of BiotechnologyKIIT University Bhubaneswar Odisha India
| | | |
Collapse
|
2
|
Nnadi CO, Ebiloma GU, Black JA, Nwodo NJ, Lemgruber L, Schmidt TJ, de Koning HP. Potent Antitrypanosomal Activities of 3-Aminosteroids against African Trypanosomes: Investigation of Cellular Effects and of Cross-Resistance with Existing Drugs. Molecules 2019; 24:E268. [PMID: 30642032 PMCID: PMC6359104 DOI: 10.3390/molecules24020268] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 11/16/2022] Open
Abstract
Treatment of animal African trypanosomiasis (AAT) requires urgent need for safe, potent and affordable drugs and this has necessitated this study. We investigated the trypanocidal activities and mode of action of selected 3-aminosteroids against Trypanosoma brucei brucei. The in vitro activity of selected compounds of this series against T. congolense (Savannah-type, IL3000), T. b. brucei (bloodstream trypomastigote, Lister strain 427 wild-type (427WT)) and various multi-drug resistant cell lines was assessed using a resazurin-based cell viability assay. Studies on mode of antitrypanosomal activity of some selected 3-aminosteroids against Tbb 427WT were also carried out. The tested compounds mostly showed moderate-to-low in vitro activities and low selectivity to mammalian cells. Interestingly, a certain aminosteroid, holarrhetine (10, IC50 = 0.045 ± 0.03 µM), was 2 times more potent against T. congolense than the standard veterinary drug, diminazene aceturate, and 10 times more potent than the control trypanocide, pentamidine, and displayed an excellent in vitro selectivity index of 2130 over L6 myoblasts. All multi-drug resistant strains of T. b. brucei tested were not significantly cross-resistant with the purified compounds. The growth pattern of Tbb 427WT on long and limited exposure time revealed gradual but irrecoverable growth arrest at ≥ IC50 concentrations of 3-aminosteroids. Trypanocidal action was not associated with membrane permeabilization of trypanosome cells but instead with mitochondrial membrane depolarization, reduced adenosine triphosphate (ATP) levels and G₂/M cell cycle arrest which appear to be the result of mitochondrial accumulation of the aminosteroids. These findings provided insights for further development of this new and promising class of trypanocide against African trypanosomes.
Collapse
Affiliation(s)
- Charles O Nnadi
- Institute of Pharmaceutical Biology and Phytochemistry (IPBP), University of Münster, Pharma Campus Corrensstraße 48, D-48149 Münster, Germany.
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria Nsukka, Enugu 410001, Nigeria.
| | - Godwin U Ebiloma
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK.
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto 606-8585, Japan.
| | - Jennifer A Black
- The Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK.
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil.
| | - Ngozi J Nwodo
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria Nsukka, Enugu 410001, Nigeria.
| | - Leandro Lemgruber
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK.
| | - Thomas J Schmidt
- Institute of Pharmaceutical Biology and Phytochemistry (IPBP), University of Münster, Pharma Campus Corrensstraße 48, D-48149 Münster, Germany.
| | - Harry P de Koning
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK.
| |
Collapse
|
3
|
Das P, Babbar P, Malhotra N, Sharma M, Jachak GR, Gonnade RG, Shanmugam D, Harlos K, Yogavel M, Sharma A, Reddy DS. Specific Stereoisomeric Conformations Determine the Drug Potency of Cladosporin Scaffold against Malarial Parasite. J Med Chem 2018; 61:5664-5678. [DOI: 10.1021/acs.jmedchem.8b00565] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Pronay Das
- Organic Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - Palak Babbar
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Nipun Malhotra
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Manmohan Sharma
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Goraknath R. Jachak
- Organic Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - Rajesh G. Gonnade
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
- Center for Material Characterization, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
| | - Dhanasekaran Shanmugam
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
| | - Karl Harlos
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, The Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Manickam Yogavel
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Amit Sharma
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - D. Srinivasa Reddy
- Organic Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| |
Collapse
|
4
|
Voorberg-van der Wel A, Roma G, Gupta DK, Schuierer S, Nigsch F, Carbone W, Zeeman AM, Lee BH, Hofman SO, Faber BW, Knehr J, Pasini E, Kinzel B, Bifani P, Bonamy GMC, Bouwmeester T, Kocken CHM, Diagana TT. A comparative transcriptomic analysis of replicating and dormant liver stages of the relapsing malaria parasite Plasmodium cynomolgi. eLife 2017; 6:29605. [PMID: 29215331 PMCID: PMC5758109 DOI: 10.7554/elife.29605] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
Plasmodium liver hypnozoites, which cause disease relapse, are widely considered to be the last barrier towards malaria eradication. The biology of this quiescent form of the parasite is poorly understood which hinders drug discovery. We report a comparative transcriptomic dataset of replicating liver schizonts and dormant hypnozoites of the relapsing parasite Plasmodium cynomolgi. Hypnozoites express only 34% of Plasmodium physiological pathways, while 91% are expressed in replicating schizonts. Few known malaria drug targets are expressed in quiescent parasites, but pathways involved in microbial dormancy, maintenance of genome integrity and ATP homeostasis were robustly expressed. Several transcripts encoding heavy metal transporters were expressed in hypnozoites and the copper chelator neocuproine was cidal to all liver stage parasites. This transcriptomic dataset is a valuable resource for the discovery of vaccines and effective treatments to combat vivax malaria.
Collapse
Affiliation(s)
| | - Guglielmo Roma
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Sven Schuierer
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Florian Nigsch
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Walter Carbone
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Boon Heng Lee
- Novartis Institute for Tropical Diseases, Singapore, Singapore
| | - Sam O Hofman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Judith Knehr
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Erica Pasini
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Bernd Kinzel
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Pablo Bifani
- Novartis Institute for Tropical Diseases, Singapore, Singapore
| | | | | | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | | |
Collapse
|
5
|
Varela JN, Lammoglia Cobo MF, Pawar SV, Yadav VG. Cheminformatic Analysis of Antimalarial Chemical Space Illuminates Therapeutic Mechanisms and Offers Strategies for Therapy Development. J Chem Inf Model 2017; 57:2119-2131. [PMID: 28810125 DOI: 10.1021/acs.jcim.7b00072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The clear and present danger of malaria, which has been amplified in recent years by climate change, and the progressive thinning of our drug arsenal over the past two decades raise uncomfortable questions about the current state and future of antimalarial drug development. Besides suffering from many of the same technical challenges that affect drug development in other disease areas, the quest for new antimalarial therapies is also hindered by the complex, dynamic life cycle of the malaria parasite, P. falciparum, in its mosquito and human hosts, and its role thereof in the elicitation of drug resistance. New strategies are needed in order to ensure economical and expeditious development of new, more efficacious treatments. In the present study, we employ open-source cheminformatics tools to analyze the chemical space traversed by approved antimalarial drugs and promising candidates at various stages of development to uncover insights that could shape future endeavors in the field. Our scaffold-centric analysis reveals that the antimalarial chemical space is disjointed and segregated into a few dominant structural groups. In fact, the structures of antimalarial drugs and drug candidates are distributed according to Pareto's principle. This structural convergence can potentially be exploited for future drug discovery by incorporating it into bioinformatics workflows that are typically employed for solving problems in structural biology. Significantly, we demonstrate how molecular scaffold hunting can be applied to unearth putative mechanisms of action of drugs whose activities remain a mystery, and how scaffold-centric analysis of drug space can also provide a recipe for combination therapies that minimize the likelihood of emergence of drug resistance, as well as identify areas on which to focus efforts. Finally, we also observe that over half of the molecules in the antimalarial space bear no resemblance to other molecules in the collection, which suggests that the pharmacobiology of antimalarial drugs has not been entirely surveyed.
Collapse
Affiliation(s)
- Julia Nogueira Varela
- Department of Chemical & Biological Engineering, The University of British Columbia , Vancouver, BC, Canada , V6T 1Z3
| | - María Fernanda Lammoglia Cobo
- Department of Chemical & Biological Engineering, The University of British Columbia , Vancouver, BC, Canada , V6T 1Z3.,Life Sciences Department, Monterrey Institute of Technology and Higher Education , Mexico City Campus, Mexico City, Mexico , 14380
| | - Sandip V Pawar
- Department of Chemical & Biological Engineering, The University of British Columbia , Vancouver, BC, Canada , V6T 1Z3
| | - Vikramaditya G Yadav
- Department of Chemical & Biological Engineering, The University of British Columbia , Vancouver, BC, Canada , V6T 1Z3.,Neglected Global Diseases Initiative, The University of British Columbia , Vancouver, BC, Canada , V6T 1Z3
| |
Collapse
|
6
|
Edwards RL, Odom John AR. Muddled mechanisms: recent progress towards antimalarial target identification. F1000Res 2016; 5:2514. [PMID: 27803804 PMCID: PMC5070598 DOI: 10.12688/f1000research.9477.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/06/2016] [Indexed: 01/06/2023] Open
Abstract
In the past decade, malaria rates have plummeted as a result of aggressive infection control measures and the adoption of artemisinin-based combination therapies (ACTs). However, a potential crisis looms ahead. Treatment failures to standard antimalarial regimens have been reported in Southeast Asia, and devastating consequences are expected if resistance spreads to the African continent. To prevent a potential public health emergency, the antimalarial arsenal must contain therapeutics with novel mechanisms of action (MOA). An impressive number of high-throughput screening (HTS) campaigns have since been launched, identifying thousands of compounds with activity against one of the causative agents of malaria,
Plasmodium falciparum. Now begins the difficult task of target identification, for which studies are often tedious, labor intensive, and difficult to interpret. In this review, we highlight approaches that have been instrumental in tackling the challenges of target assignment and elucidation of the MOA for hit compounds. Studies that apply these innovative techniques to antimalarial target identification are described, as well as the impact of the data in the field.
Collapse
Affiliation(s)
- Rachel L Edwards
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Audrey R Odom John
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
7
|
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: 1.7] [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]
|
8
|
Shamriz S, Ofoghi H. Design, structure prediction and molecular dynamics simulation of a fusion construct containing malaria pre-erythrocytic vaccine candidate, PfCelTOS, and human interleukin 2 as adjuvant. BMC Bioinformatics 2016; 17:71. [PMID: 26851942 PMCID: PMC4744421 DOI: 10.1186/s12859-016-0918-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/29/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Malaria infection is still widespread in some parts of the world and threatens the lives of millions of people every year. Vaccines, especially oral vaccines are considered to be effective in reducing the burden of malaria morbidity and mortality. By using recombinant technology, suitable oral hosts could serve as antigen delivering vehicles in developing oral vaccines. This study was aimed towards designing and computational analysis of a fusion protein consisting of Plasmodium falciparum cell-traversal protein for ookinetes and sporozoites (PfCelTOS) fused to human interleukin-2 (IL-2) and M cell-specific peptide ligand (Co1), as a step toward developing a vaccine candidate. RESULTS To our best knowledge, the three dimensional (3D) structure of CelTOS is not reported in protein database. Therefore, we carried out computational modeling and simulation in the hope of understanding the properties and structure of PfCelTOS. Then we fused IL-2 to PfCelTOS by a flexible linker and did in silico analysis to confirm the proper folding of each domain in the designed fusion protein. In the last step, Co1 ligand was added to the confirmed fusion structure using a rigid linker and computational analysis was performed to evaluate the final fusion construct. One structure out of five predicted by I-TASSER for PfCelTOS and fusion constructs was selected based on the highest value for C-score. Molecular dynamics (MD) simulation analysis indicated that predicted structures are stable during the simulation. Ramchandran Plot analysis of PfCelTOS and fusion constructs before and after MD simulation also represented that most residues were fallen in favorable regions. CONCLUSION In silico study showed that Co1-(AEEEK)3- IL-2-(GGGGS)3-PfCelTOS construct has a constant structure and the selected linkers are effectively able to separate the domains. Therefore, data reported in this paper represents the first step toward developing of an oral vaccine candidate against malaria infection.
Collapse
Affiliation(s)
- Shabnam Shamriz
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran.
| | - Hamideh Ofoghi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran.
| |
Collapse
|
9
|
Siwo GH, Smith RS, Tan A, Button-Simons KA, Checkley LA, Ferdig MT. An integrative analysis of small molecule transcriptional responses in the human malaria parasite Plasmodium falciparum. BMC Genomics 2015; 16:1030. [PMID: 26637195 PMCID: PMC4670519 DOI: 10.1186/s12864-015-2165-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 10/29/2015] [Indexed: 12/05/2022] Open
Abstract
Background Transcriptional responses to small molecules can provide insights into drug mode of action (MOA). The capacity of the human malaria parasite, Plasmodium falciparum, to respond specifically to transcriptional perturbations has been unclear based on past approaches. Here, we present the most extensive profiling to date of the parasite’s transcriptional responsiveness to thirty-one chemically and functionally diverse small molecules. Methods We exposed two laboratory strains of the human malaria parasite P. falciparum to brief treatments of thirty-one chemically and functionally diverse small molecules associated with biological effects across multiple pathways based on various levels of evidence. We investigated the impact of chemical composition and MOA on gene expression similarities that arise between perturbations by various compounds. To determine the target biological pathways for each small molecule, we developed a novel framework for encoding small molecule effects on a spectra of biological processes or GO functions that are enriched in the differentially expressed genes of a given small molecule perturbation. Results We find that small molecules associated with similar transcriptional responses contain similar chemical features, and/ or have a shared MOA. The approach also revealed complex relationships between drugs and biological pathways that are missed by most exisiting approaches. For example, the approach was able to partition small molecule responses into drug-specific effects versus non-specific effects. Conclusions Our work provides a new framework for linking transcriptional responses to drug MOA in P. falciparum and can be generalized for the same purpose in other organisms. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2165-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Geoffrey H Siwo
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.,Current Address: IBM TJ Watson Research Center, Yorktown Heights, NY, 10598, USA.,Current Address: IBM Research-Africa, South Africa Lab, Sandton, Johannesburg, 2196, South Africa
| | - Roger S Smith
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.,Current Address: Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Asako Tan
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.,Epicenter, Madison, WI, 53719, USA
| | - Katrina A Button-Simons
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Lisa A Checkley
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Michael T Ferdig
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
10
|
New paradigm of an old target: an update on structural biology and current progress in drug design towards plasmepsin II. Eur J Med Chem 2015; 95:324-48. [PMID: 25827401 DOI: 10.1016/j.ejmech.2015.03.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/09/2015] [Accepted: 03/20/2015] [Indexed: 11/20/2022]
Abstract
Malaria is one of the major parasitic disease whose rapid spreading and mortality rate affects all parts of the world especially several parts of Asia as well as Africa. The emergence of multi-drug resistant strains hamper the progress of current antimalarial therapy and displayed an urgent need for new antimalarials by targeting novel drug targets. Until now, several promising targets were explored in order to develop a promising Achilles hill to counter malaria. Plasmepsin, an aspartic protease, which is involved in the hemoglobin breakdown into smaller peptides emerged as a crucial target to develop new chemical entities to counter malaria. Due to early crystallographic evidence, plasmepsin II (Plm II) emerged as well explored target to develop novel antimalarials as well as a starting point to develop inhibitors targeting some other subtypes of plasmepsins i.e. Plm I, II, IV and V. With the advancements in drug discovery, several computational and synthetic approaches were employed in order to develop novel inhibitors targeting Plm II. Strategies such as fragment based drug design, molecular dynamics simulation, double drug approach etc. were employed in order to develop new chemical entities targeting Plm II. But majority of Plm II inhibitors suffered from poor selectivity over cathepsin D as well as other subtypes of plasmepsins. This review highlights an updated account of drug discovery efforts targeting plasmepsin II from a medicinal chemistry perspective.
Collapse
|
11
|
Abstract
Despite a century of control and eradication campaigns, malaria remains one of the world's most devastating diseases. Our once-powerful therapeutic weapons are losing the war against the Plasmodium parasite, whose ability to rapidly develop and spread drug resistance hamper past and present malaria-control efforts. Finding new and effective treatments for malaria is now a top global health priority, fuelling an increase in funding and promoting open-source collaborations between researchers and pharmaceutical consortia around the world. The result of this is rapid advances in drug discovery approaches and technologies, with three major methods for antimalarial drug development emerging: (i) chemistry-based, (ii) target-based, and (iii) cell-based. Common to all three of these approaches is the unique ability of structural biology to inform and accelerate drug development. Where possible, SBDD (structure-based drug discovery) is a foundation for antimalarial drug development programmes, and has been invaluable to the development of a number of current pre-clinical and clinical candidates. However, as we expand our understanding of the malarial life cycle and mechanisms of resistance development, SBDD as a field must continue to evolve in order to develop compounds that adhere to the ideal characteristics for novel antimalarial therapeutics and to avoid high attrition rates pre- and post-clinic. In the present review, we aim to examine the contribution that SBDD has made to current antimalarial drug development efforts, covering hit discovery to lead optimization and prevention of parasite resistance. Finally, the potential for structural biology, particularly high-throughput structural genomics programmes, to identify future targets for drug discovery are discussed.
Collapse
|
12
|
KAF156 is an antimalarial clinical candidate with potential for use in prophylaxis, treatment, and prevention of disease transmission. Antimicrob Agents Chemother 2014; 58:5060-7. [PMID: 24913172 PMCID: PMC4135840 DOI: 10.1128/aac.02727-13] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Renewed global efforts toward malaria eradication have highlighted the need for novel antimalarial agents with activity against multiple stages of the parasite life cycle. We have previously reported the discovery of a novel class of antimalarial compounds in the imidazolopiperazine series that have activity in the prevention and treatment of blood stage infection in a mouse model of malaria. Consistent with the previously reported activity profile of this series, the clinical candidate KAF156 shows blood schizonticidal activity with 50% inhibitory concentrations of 6 to 17.4 nM against P. falciparum drug-sensitive and drug-resistant strains, as well as potent therapeutic activity in a mouse models of malaria with 50, 90, and 99% effective doses of 0.6, 0.9, and 1.4 mg/kg, respectively. When administered prophylactically in a sporozoite challenge mouse model, KAF156 is completely protective as a single oral dose of 10 mg/kg. Finally, KAF156 displays potent Plasmodium transmission blocking activities both in vitro and in vivo. Collectively, our data suggest that KAF156, currently under evaluation in clinical trials, has the potential to treat, prevent, and block the transmission of malaria.
Collapse
|
13
|
Biamonte MA, Wanner J, Le Roch KG. Recent advances in malaria drug discovery. Bioorg Med Chem Lett 2013; 23:2829-43. [PMID: 23587422 PMCID: PMC3762334 DOI: 10.1016/j.bmcl.2013.03.067] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/11/2013] [Accepted: 03/20/2013] [Indexed: 01/18/2023]
Abstract
This digest covers some of the most relevant progress in malaria drug discovery published between 2010 and 2012. There is an urgent need to develop new antimalarial drugs. Such drugs can target the blood stage of the disease to alleviate the symptoms, the liver stage to prevent relapses, and the transmission stage to protect other humans. The pipeline for the blood stage is becoming robust, but this should not be a source of complacency, as the current therapies set a high standard. Drug discovery efforts directed towards the liver and transmission stages are in their infancy but are receiving increasing attention as targeting these stages could be instrumental in eradicating malaria.
Collapse
Affiliation(s)
- Marco A Biamonte
- Drug Discovery for Tropical Diseases, Suite 230, San Diego, CA 92121, USA.
| | | | | |
Collapse
|
14
|
Rahul CN, Shiva Krishna K, Pawar AP, Bai M, Kumar V, Phadke S, Rajesh V. Genetic and structural characterization of PvSERA4: potential implication as therapeutic target for Plasmodium vivax malaria. J Biomol Struct Dyn 2013; 32:580-90. [PMID: 23582016 DOI: 10.1080/07391102.2013.782824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Plasmodium vivax malaria is geographically the most widely distributed and prevalent form of human malaria. The development of drug resistance by the parasite to existing drugs necessitates higher focus to explore and identify new drug targets. Plasmodial proteases have key roles in parasite biology and are involved in nutritional uptake, egress from infected reticulocytes, and invasion of the new target erythrocytes. Serine repeat antigens (SERA) of Plasmodium are parasite proteases that remain attractive drug targets and are important vaccine candidates due to their high expression profiles in the blood stages. SERA proteins have a unique putative papain-like cysteine protease motif that has either serine or cysteine in its active site. In P. vivax, PvSERA4 is the highest transcribed member of this multigene family. In this study, we have investigated the genetic polymorphism of PvSERA4 central protease domain and deduced its 3D model by homology modeling and also performed MD simulations to acquire refined protein structure. Sequence analysis of protease domain of PvSERA4 from Indian field isolates reveals that the central domain is highly conserved. The high sequence conservation of the PvSERA4 enzyme domain coupled with its high expression raises the possibility of it having a critical role in parasite biology and hence, being a reliable target for new selective inhibitor-based antimalarial chemotherapeutics. The 3D model showed the presence of an unusual antiparallel Beta hairpin motif between catalytic residues similar to hemoglobin binding motif of Plasmodial hemoglobinases. Our PvSERA4 model will aid in designing structure-based inhibitors against this enzyme.
Collapse
Affiliation(s)
- C N Rahul
- a Department of Biological Sciences , Birla Institute of Technology and Science, Pilani , Hyderabad Campus , Andhra Pradesh , India
| | | | | | | | | | | | | |
Collapse
|
15
|
Schenone M, Dančík V, Wagner BK, Clemons PA. Target identification and mechanism of action in chemical biology and drug discovery. Nat Chem Biol 2013; 9:232-40. [PMID: 23508189 PMCID: PMC5543995 DOI: 10.1038/nchembio.1199] [Citation(s) in RCA: 697] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/28/2013] [Indexed: 12/12/2022]
Abstract
Target-identification and mechanism-of-action studies have important roles in small-molecule probe and drug discovery. Biological and technological advances have resulted in the increasing use of cell-based assays to discover new biologically active small molecules. Such studies allow small-molecule action to be tested in a more disease-relevant setting at the outset, but they require follow-up studies to determine the precise protein target or targets responsible for the observed phenotype. Target identification can be approached by direct biochemical methods, genetic interactions or computational inference. In many cases, however, combinations of approaches may be required to fully characterize on-target and off-target effects and to understand mechanisms of small-molecule action.
Collapse
Affiliation(s)
- Monica Schenone
- Proteomics Platform, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Vlado Dančík
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Bridget K Wagner
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Paul A Clemons
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
16
|
Riley EM, Stewart VA. Immune mechanisms in malaria: new insights in vaccine development. Nat Med 2013; 19:168-78. [DOI: 10.1038/nm.3083] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
|
17
|
Hoepfner D, McNamara CW, Lim CS, Studer C, Riedl R, Aust T, McCormack SL, Plouffe DM, Meister S, Schuierer S, Plikat U, Hartmann N, Staedtler F, Cotesta S, Schmitt EK, Petersen F, Supek F, Glynne RJ, Tallarico JA, Porter JA, Fishman MC, Bodenreider C, Diagana TT, Movva NR, Winzeler EA. Selective and specific inhibition of the plasmodium falciparum lysyl-tRNA synthetase by the fungal secondary metabolite cladosporin. Cell Host Microbe 2012; 11:654-63. [PMID: 22704625 PMCID: PMC3391680 DOI: 10.1016/j.chom.2012.04.015] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/02/2012] [Accepted: 04/22/2012] [Indexed: 01/06/2023]
Abstract
With renewed calls for malaria eradication, next-generation antimalarials need be active against drug-resistant parasites and efficacious against both liver- and blood-stage infections. We screened a natural product library to identify inhibitors of Plasmodium falciparum blood- and liver-stage proliferation. Cladosporin, a fungal secondary metabolite whose target and mechanism of action are not known for any species, was identified as having potent, nanomolar, antiparasitic activity against both blood and liver stages. Using postgenomic methods, including a yeast deletion strains collection, we show that cladosporin specifically inhibits protein synthesis by directly targeting P. falciparum cytosolic lysyl-tRNA synthetase. Further, cladosporin is >100-fold more potent against parasite lysyl-tRNA synthetase relative to the human enzyme, which is conferred by the identity of two amino acids within the enzyme active site. Our data indicate that lysyl-tRNA synthetase is an attractive, druggable, antimalarial target that can be selectively inhibited.
Collapse
Affiliation(s)
- Dominic Hoepfner
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum 1 Novartis Campus, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chatterjee AK, Yeung BKS. Back to the future: lessons learned in modern target-based and whole-cell lead optimization of antimalarials. Curr Top Med Chem 2012; 12:473-83. [PMID: 22242845 PMCID: PMC3355380 DOI: 10.2174/156802612799362977] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 11/09/2011] [Accepted: 11/17/2011] [Indexed: 11/22/2022]
Abstract
Antimalarial drug discovery has historically benefited from the whole-cell (phenotypic) screening approach to identify lead molecules in the search for new drugs. However over the past two decades there has been a shift in the pharmaceutical industry to move away from whole-cell screening to target-based approaches. As part of a Wellcome Trust and Medicines for Malaria Venture (MMV) funded consortium to discover new blood-stage antimalarials, we used both approaches to identify new antimalarial chemotypes, two of which have progressed beyond the lead optimization phase and display excellent in vivo efficacy in mice. These two advanced series were identified through a cell-based optimization devoid of target information and in this review we summarize the advantages of this approach versus a target-based optimization. Although the each lead optimization required slightly different medicinal chemistry strategies, we observed some common issues across the different the scaffolds which could be applied to other cell based lead optimization programs.
Collapse
Affiliation(s)
- Arnab K Chatterjee
- Genomics Institute, Novartis Research Foundation, San Diego, CA 92121, USA.
| | | |
Collapse
|
19
|
Guiguemde WA, Shelat AA, Garcia-Bustos JF, Diagana TT, Gamo FJ, Guy RK. Global phenotypic screening for antimalarials. ACTA ACUST UNITED AC 2012; 19:116-29. [PMID: 22284359 DOI: 10.1016/j.chembiol.2012.01.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/10/2012] [Accepted: 01/10/2012] [Indexed: 12/20/2022]
Abstract
Malaria, a devastating infectious disease caused by Plasmodium spp., leads to roughly 655,000 deaths per year, mostly of African children. To compound the problem, drug resistance has emerged to all classical antimalarials and may be emerging for artemisinin-based combination therapies. To address the need for new antimalarials with novel mechanisms, several groups carried out phenotypic screening campaigns to identify compounds inhibiting growth of the blood stages of Plasmodium falciparum. In this review, we describe the characterization of these compounds, explore currently ongoing strategies to develop lead molecules, and endorse the concept of a "malaria box" of publicly accessible active compounds.
Collapse
Affiliation(s)
- W Armand Guiguemde
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Malaria is an important human disease and is the target of a global eradication campaign. New technological and informatics advancements in population genomics are being leveraged to identify genetic loci under selection in the malaria parasite and to find variants that are associated with key clinical phenotypes, such as drug resistance. This article provides a timely Review of how population-genetics-based strategies are being applied to Plasmodium falciparum both to identify genetic loci as key targets of interventions and to develop monitoring and surveillance tools that are crucial for the successful elimination and eradication of malaria.
Collapse
|
21
|
Application of genomics to field investigations of malaria by the international centers of excellence for malaria research. Acta Trop 2012; 121:324-32. [PMID: 22182668 DOI: 10.1016/j.actatropica.2011.12.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 11/28/2011] [Accepted: 12/02/2011] [Indexed: 12/15/2022]
Abstract
Success of the global research agenda toward eradication of malaria will depend on development of new tools, including drugs, vaccines, insecticides and diagnostics. Genomic information, now available for the malaria parasites, their mosquito vectors, and human host, can be leveraged to both develop these tools and monitor their effectiveness. Although knowledge of genomic sequences for the malaria parasites, Plasmodium falciparum and Plasmodium vivax, have helped advance our understanding of malaria biology, simply knowing this sequence information has not yielded a plethora of new interventions to reduce the burden of malaria. Here we review and provide specific examples of how genomic information has increased our knowledge of parasite biology, focusing on P. falciparum malaria. We then discuss how population genetics can be applied toward the epidemiological and transmission-related goals outlined by the International Centers of Excellence for Malaria Research groups recently established by the National Institutes of Health. Finally, we propose genomics is a research area that can promote coordination and collaboration between various ICEMR groups, and that working together as a community can significantly advance the value of this information toward reduction of the global malaria burden.
Collapse
|
22
|
Heidebrecht RW, Mulrooney C, Austin CP, Barker RH, Beaudoin JA, Cheng KCC, Comer E, Dandapani S, Dick J, Duvall JR, Ekland EH, Fidock DA, Fitzgerald M, Foley M, Guha R, Hinkson P, Kramer M, Lukens AK, Masi D, Marcaurelle L, Su XZ, Thomas CJ, Weïwer M, Wiegand RC, Wirth D, Xia M, Yuan J, Zhao J, Palmer M, Munoz B, Schreiber S. Diversity-Oriented Synthesis Yields a Novel Lead for the Treatment of Malaria. ACS Med Chem Lett 2012; 3:112-117. [PMID: 22328964 PMCID: PMC3276110 DOI: 10.1021/ml200244k] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 12/14/2011] [Indexed: 02/06/2023] Open
Abstract
Here, we describe the discovery of a novel antimalarial agent using phenotypic screening of Plasmodium falciparum asexual blood-stage parasites. Screening a novel compound collection created using diversity-oriented synthesis (DOS) led to the initial hit. Structure-activity relationships guided the synthesis of compounds having improved potency and water solubility, yielding a subnanomolar inhibitor of parasite asexual blood-stage growth. Optimized compound 27 has an excellent off-target activity profile in erythrocyte lysis and HepG2 assays and is stable in human plasma. This compound is available via the molecular libraries probe production centers network (MLPCN) and is designated ML238.
Collapse
Affiliation(s)
- Richard W. Heidebrecht
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
- Harvard School of Public Health, Huntington Avenue,
Boston, Massachusetts 02115, United States
| | - Carol Mulrooney
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Christopher P. Austin
- Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892,
United States
| | - Robert H. Barker
- Genzyme Corporation, 153 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Jennifer A. Beaudoin
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Ken Chih-Chien Cheng
- Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892,
United States
| | - Eamon Comer
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Sivaraman Dandapani
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Justin Dick
- Harvard School of Public Health, Huntington Avenue,
Boston, Massachusetts 02115, United States
| | - Jeremy R. Duvall
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Eric H. Ekland
- Department of Microbiology and Immunology, Colombia University, New York, New York 10032, United
States
| | - David A. Fidock
- Department of Microbiology and Immunology, Colombia University, New York, New York 10032, United
States
- Department of Medicine,
Division of Infectious Diseases, Colombia University, New York, New York 10032, United States
| | - Mark
E. Fitzgerald
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Michael Foley
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Rajarshi Guha
- Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892,
United States
| | - Paul Hinkson
- Genzyme Corporation, 153 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Martin Kramer
- Genzyme Corporation, 153 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Amanda K. Lukens
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
- Harvard School of Public Health, Huntington Avenue,
Boston, Massachusetts 02115, United States
| | - Daniela Masi
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Lisa
A. Marcaurelle
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Xin-Zhuan Su
- National Institute of Allergy and Infectious
Diseases, Bethesda, Maryland 20892, United States
| | - Craig J. Thomas
- Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892,
United States
| | - Michel Weïwer
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Roger C. Wiegand
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
- Harvard School of Public Health, Huntington Avenue,
Boston, Massachusetts 02115, United States
| | - Dyann Wirth
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
- Harvard School of Public Health, Huntington Avenue,
Boston, Massachusetts 02115, United States
| | - Menghang Xia
- Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892,
United States
| | - Jing Yuan
- National Institute of Allergy and Infectious
Diseases, Bethesda, Maryland 20892, United States
| | - Jinghua Zhao
- Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892,
United States
| | - Michelle Palmer
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Benito Munoz
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Stuart Schreiber
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
- Howard Hughes Medical Institute,
Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
23
|
Abad Martínez MJ, Del Olmo LMB, Ticona LA, Benito PB. The Artemisia L. Genus. STUDIES IN NATURAL PRODUCTS CHEMISTRY VOLUME 37 2012. [DOI: 10.1016/b978-0-444-59514-0.00002-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
24
|
Nam TG, McNamara CW, Bopp S, Dharia NV, Meister S, Bonamy GMC, Plouffe DM, Kato N, McCormack S, Bursulaya B, Ke H, Vaidya AB, Schultz PG, Winzeler EA. A chemical genomic analysis of decoquinate, a Plasmodium falciparum cytochrome b inhibitor. ACS Chem Biol 2011; 6:1214-22. [PMID: 21866942 PMCID: PMC3220786 DOI: 10.1021/cb200105d] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
Decoquinate has single-digit nanomolar activity against in vitro blood stage Plasmodium falciparum parasites, the causative agent of human malaria. In vitro evolution of decoquinate-resistant parasites and subsequent comparative genomic analysis to the drug-sensitive parental strain revealed resistance was conferred by two nonsynonymous single nucleotide polymorphisms in the gene encoding cytochrome b. The resultant amino acid mutations, A122T and Y126C, reside within helix C in the ubiquinol-binding pocket of cytochrome b, an essential subunit of the cytochrome bc1 complex. As with other cytochrome bc1 inhibitors, such as atovaquone, decoquinate has low nanomolar activity against in vitro liver stage P. yoelii and provides partial prophylaxis protection when administered to infected mice at 50 mg kg–1. In addition, transgenic parasites expressing yeast dihydroorotate dehydrogenase are >200-fold less sensitive to decoquinate, which provides additional evidence that this drug inhibits the parasite’s mitochondrial electron transport chain. Importantly, decoquinate exhibits limited cross-resistance to a panel of atovaquone-resistant parasites evolved to harbor various mutations in cytochrome b. The basis for this difference was revealed by molecular docking studies, in which both of these inhibitors were shown to have distinctly different modes of binding within the ubiquinol-binding site of cytochrome b.
Collapse
Affiliation(s)
| | - Case W. McNamara
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | | | | | | | - Ghislain M. C. Bonamy
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - David M. Plouffe
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Nobutaka Kato
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Susan McCormack
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Badry Bursulaya
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Hangjun Ke
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, United States
| | - Akhil B. Vaidya
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, United States
| | - Peter G. Schultz
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Elizabeth A. Winzeler
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| |
Collapse
|
25
|
Affiliation(s)
- Alan Fairlamb
- Wellcome Principal Research Fellow, Division of Biological Chemistry & Drug Discovery, School of Life Sciences, Wellcome Trust Biocentre, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Stewart Cole
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015, Lausanne, Switzerland
| |
Collapse
|