1
|
Monteiro Júnior JC, Krüger A, Palmisano G, Wrenger C. Transporter-Mediated Solutes Uptake as Drug Target in Plasmodium falciparum. Front Pharmacol 2022; 13:845841. [PMID: 35370717 PMCID: PMC8965513 DOI: 10.3389/fphar.2022.845841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Malaria remains a public health problem with still more than half a million deaths annually. Despite ongoing efforts of many countries, malaria elimination has been difficult due to emerging resistances against most traditional drugs, including artemisinin compounds - the most potent antimalarials currently available. Therefore, the discovery and development of new drugs with novel mechanisms of action to circumvent resistances is urgently needed. In this sense, one of the most promising areas is the exploration of transport proteins. Transporters mediate solute uptake for intracellular parasite proliferation and survival. Targeting transporters can exploit these processes to eliminate the parasite. Here, we focus on transporters of the Plasmodium falciparum-infected red blood cell studied as potential biological targets and discuss published drugs directed at them.
Collapse
Affiliation(s)
- Júlio César Monteiro Júnior
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Arne Krüger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
3
|
Najmi A, Javed SA, Al Bratty M, Alhazmi HA. Modern Approaches in the Discovery and Development of Plant-Based Natural Products and Their Analogues as Potential Therapeutic Agents. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020349. [PMID: 35056662 PMCID: PMC8779633 DOI: 10.3390/molecules27020349] [Citation(s) in RCA: 197] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022]
Abstract
Natural products represents an important source of new lead compounds in drug discovery research. Several drugs currently used as therapeutic agents have been developed from natural sources; plant sources are specifically important. In the past few decades, pharmaceutical companies demonstrated insignificant attention towards natural product drug discovery, mainly due to its intrinsic complexity. Recently, technological advancements greatly helped to address the challenges and resulted in the revived scientific interest in drug discovery from natural sources. This review provides a comprehensive overview of various approaches used in the selection, authentication, extraction/isolation, biological screening, and analogue development through the application of modern drug-development principles of plant-based natural products. Main focus is given to the bioactivity-guided fractionation approach along with associated challenges and major advancements. A brief outline of historical development in natural product drug discovery and a snapshot of the prominent natural drugs developed in the last few decades are also presented. The researcher’s opinions indicated that an integrated interdisciplinary approach utilizing technological advances is necessary for the successful development of natural products. These involve the application of efficient selection method, well-designed extraction/isolation procedure, advanced structure elucidation techniques, and bioassays with a high-throughput capacity to establish druggability and patentability of phyto-compounds. A number of modern approaches including molecular modeling, virtual screening, natural product library, and database mining are being used for improving natural product drug discovery research. Renewed scientific interest and recent research trends in natural product drug discovery clearly indicated that natural products will play important role in the future development of new therapeutic drugs and it is also anticipated that efficient application of new approaches will further improve the drug discovery campaign.
Collapse
Affiliation(s)
- Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.N.); (M.A.B.); (H.A.A.)
| | - Sadique A. Javed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.N.); (M.A.B.); (H.A.A.)
- Correspondence:
| | - Mohammed Al Bratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.N.); (M.A.B.); (H.A.A.)
| | - Hassan A. Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.N.); (M.A.B.); (H.A.A.)
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
4
|
Alder A, Struck NS, Xu M, Johnson JW, Wang W, Pallant D, Cook MA, Rambow J, Lemcke S, Gilberger TW, Wright GD. A non-reactive natural product precursor of the duocarmycin family has potent and selective antimalarial activity. Cell Chem Biol 2021; 29:840-853.e6. [PMID: 34710358 DOI: 10.1016/j.chembiol.2021.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/15/2021] [Accepted: 10/02/2021] [Indexed: 11/27/2022]
Abstract
We identify a selective nanomolar inhibitor of blood-stage malarial proliferation from a screen of microbial natural product extracts. The responsible compound, PDE-I2, is a precursor of the anticancer duocarmycin family that preserves the class's sequence-specific DNA binding but lacks its signature DNA alkylating cyclopropyl warhead. While less active than duocarmycin, PDE-I2 retains comparable antimalarial potency to chloroquine. Importantly, PDE-I2 is >1,000-fold less toxic to human cell lines than duocarmycin, with mitigated impacts on eukaryotic chromosome stability. PDE-I2 treatment induces severe defects in parasite nuclear segregation leading to impaired daughter cell formation during schizogony. Time-of-addition studies implicate parasite DNA metabolism as the target of PDE-I2, with defects observed in DNA replication and chromosome integrity. We find the effect of duocarmycin and PDE-I2 on parasites is phenotypically indistinguishable, indicating that the DNA binding specificity of duocarmycins is sufficient and the genotoxic cyclopropyl warhead is dispensable for the parasite-specific selectivity of this compound class.
Collapse
Affiliation(s)
- Arne Alder
- Centre for Structural Systems Biology, 22607 Hamburg, Germany; Bernhard Nocht Institute for Tropical Medicine, Department of Cellular Parasitology, 20359 Hamburg, Germany; University of Hamburg, Department of Biology, 20146 Hamburg, Germany
| | - Nicole S Struck
- Bernhard Nocht Institute for Tropical Medicine, Department of Cellular Parasitology, 20359 Hamburg, Germany; M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; German Centre for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Min Xu
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jarrod W Johnson
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Wenliang Wang
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Daniel Pallant
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Michael A Cook
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Janis Rambow
- Centre for Structural Systems Biology, 22607 Hamburg, Germany; Bernhard Nocht Institute for Tropical Medicine, Department of Cellular Parasitology, 20359 Hamburg, Germany; University of Hamburg, Department of Biology, 20146 Hamburg, Germany
| | - Sarah Lemcke
- Centre for Structural Systems Biology, 22607 Hamburg, Germany; Bernhard Nocht Institute for Tropical Medicine, Department of Cellular Parasitology, 20359 Hamburg, Germany; University of Hamburg, Department of Biology, 20146 Hamburg, Germany
| | - Tim W Gilberger
- Centre for Structural Systems Biology, 22607 Hamburg, Germany; Bernhard Nocht Institute for Tropical Medicine, Department of Cellular Parasitology, 20359 Hamburg, Germany; University of Hamburg, Department of Biology, 20146 Hamburg, Germany.
| | - Gerard D Wright
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
5
|
Ayankunle AA, Wakeel OK, Kolawole OT, Oyekale AO, Ojurongbe O, Adeyeba OA. Drug Repositioning: Antimalarial Activities of GABA Analogs in Mice Infected with Plasmodium berghei. Cent Nerv Syst Agents Med Chem 2021; 20:110-121. [PMID: 32496991 DOI: 10.2174/1871524920666200604151907] [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: 12/18/2019] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Drug repositioning is becoming popular due to the development of resistance to almost all the recommended antimalarials. Pregabalin and gabapentin are chemical analogs of gamma- aminobutyric acid (GABA) approved for the treatment of epilepsy and neuropathic pain. OBJECTIVE This study investigates acute toxicities and antimalarial activities of pregabalin and gabapentin in the murine malarial model. METHODS Acute toxicities were assessed using the method of Lorke, while curative activities were assessed by the administration of serial doses of pregabalin and gabapentin to Plasmodium berghei infected mice. Pregabalin was further investigated for its prophylactic activity, and curative potential when combined with either artesunate or amodiaquine. All drugs were freshly prepared and administered orally. Thin films were collected, stained, and observed under the microscope for the estimation of parasitemia and calculation of percentage chemoinhibition or chemoprevention. In pregabalin -artesunate or -amodiaquine combination aspect of this study, survival day post-infection (SDPI) was recorded, while parasitemia was re-estimated for animals that survived till day 28. RESULTS The oral LD50 of gabapentin, as well as pregabalin, was >5,000 mg/kg. Gabapentin at 100 and 200 mg/Kg demonstrated 35.64% and -12.78% chemoinhibition, respectively, while pregabalin demonstrated 75.60% and 100.00% chemoinhibition at doses of 12.5 and 25 mg/Kg, respectively. Moreover, pregabalin at individual doses of 25, 50 mg/Kg, and in combination with either artesunate or amodiaquine demonstrated 100.00% chemoinhibition. In its prophylactic study, pregabalin was found to be 100% chemopreventive at individual doses of 12.5 and 25 mg/Kg. CONCLUSION Both GABA analogs have antimalarial properties, but pregabalin proved to be more efficacious.
Collapse
Affiliation(s)
- Akeem A Ayankunle
- Department of Pharmacology and Therapeutics, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Olayemi K Wakeel
- Department of Pharmacology and Therapeutics, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oyetunji T Kolawole
- Department of Pharmacology and Therapeutics, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Adesola O Oyekale
- Department of Chemical Pathology, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Olusola Ojurongbe
- Department of Medical Microbiology & Parasitology, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oluwaseyi A Adeyeba
- Department of Medical Microbiology & Parasitology, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
6
|
Abstract
It is estimated that more than 1 billion people across the world are affected by a neglected tropical disease (NTD) that requires medical intervention. These diseases tend to afflict people in areas with high rates of poverty and cost economies billions of dollars every year. Collaborative drug discovery efforts are required to reduce the burden of these diseases in endemic regions. The release of “Open Access Boxes” is an initiative launched by Medicines for Malaria Venture (MMV) in collaboration with its partners to catalyze new drug discovery in neglected diseases. These boxes are mainly requested by biology researchers across the globe who may not otherwise have access to compounds to screen nor knowledge of the workflow that needs to be followed after identification of actives from their screening campaigns. Here, we present guidelines on how to move such actives beyond the hit identification stage, to help in capacity strengthening and enable a greater impact of the initiative.
Collapse
|
7
|
Ferreira LT, Borba JVB, Moreira-Filho JT, Rimoldi A, Andrade CH, Costa FTM. QSAR-Based Virtual Screening of Natural Products Database for Identification of Potent Antimalarial Hits. Biomolecules 2021; 11:biom11030459. [PMID: 33808643 PMCID: PMC8003391 DOI: 10.3390/biom11030459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 01/15/2023] Open
Abstract
With about 400,000 annual deaths worldwide, malaria remains a public health burden in tropical and subtropical areas, especially in low-income countries. Selection of drug-resistant Plasmodium strains has driven the need to explore novel antimalarial compounds with diverse modes of action. In this context, biodiversity has been widely exploited as a resourceful channel of biologically active compounds, as exemplified by antimalarial drugs such as quinine and artemisinin, derived from natural products. Thus, combining a natural product library and quantitative structure-activity relationship (QSAR)-based virtual screening, we have prioritized genuine and derivative natural compounds with potential antimalarial activity prior to in vitro testing. Experimental validation against cultured chloroquine-sensitive and multi-drug-resistant P. falciparum strains confirmed the potent and selective activity of two sesquiterpene lactones (LDT-597 and LDT-598) identified in silico. Quantitative structure-property relationship (QSPR) models predicted absorption, distribution, metabolism, and excretion (ADME) and physiologically based pharmacokinetic (PBPK) parameters for the most promising compound, showing that it presents good physiologically based pharmacokinetic properties both in rats and humans. Altogether, the in vitro parasite growth inhibition results obtained from in silico screened compounds encourage the use of virtual screening campaigns for identification of promising natural compound-based antimalarial molecules.
Collapse
Affiliation(s)
- Letícia Tiburcio Ferreira
- Laboratory of Tropical Diseases Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP 13083-864, Brazil; (L.T.F.); (J.V.B.B.); (A.R.)
| | - Joyce V. B. Borba
- Laboratory of Tropical Diseases Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP 13083-864, Brazil; (L.T.F.); (J.V.B.B.); (A.R.)
- Laboratory of Molecular Modeling and Drug Design, LabMol, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO 74605-170, Brazil; (J.T.M.-F.); (C.H.A.)
| | - José Teófilo Moreira-Filho
- Laboratory of Molecular Modeling and Drug Design, LabMol, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO 74605-170, Brazil; (J.T.M.-F.); (C.H.A.)
| | - Aline Rimoldi
- Laboratory of Tropical Diseases Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP 13083-864, Brazil; (L.T.F.); (J.V.B.B.); (A.R.)
| | - Carolina Horta Andrade
- Laboratory of Molecular Modeling and Drug Design, LabMol, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO 74605-170, Brazil; (J.T.M.-F.); (C.H.A.)
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP 13083-864, Brazil; (L.T.F.); (J.V.B.B.); (A.R.)
- Correspondence: ; Tel.: +55-19-3521-6288
| |
Collapse
|
8
|
Xu Z, Shen C, Zhang H, Wang P, Dong K. Constructing chiral aza-quaternary carbon centers by enantioselective carbonylative Heck reaction of o-iodoanilines with allenes. Org Chem Front 2021. [DOI: 10.1039/d0qo01486a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The construction of chiral aza-quaternary C-centers via C–N bond formation is achieved by a Pd-catalysed asymmetric carbonylative Heck reaction of o-iodoanilines with allenes, providing chiral dihydroquinolinone derivatives with moderate to high yield and enantiomeric ratio.
Collapse
Affiliation(s)
- Zhengshuai Xu
- Chang-Kung Chuang Institute
- Shanghai Key Laboratory of Green Chemistry and Chemical Process
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| | - Chaoren Shen
- Chang-Kung Chuang Institute
- Shanghai Key Laboratory of Green Chemistry and Chemical Process
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| | - Hongru Zhang
- Chang-Kung Chuang Institute
- Shanghai Key Laboratory of Green Chemistry and Chemical Process
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| | - Peng Wang
- Chang-Kung Chuang Institute
- Shanghai Key Laboratory of Green Chemistry and Chemical Process
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| | - Kaiwu Dong
- Chang-Kung Chuang Institute
- Shanghai Key Laboratory of Green Chemistry and Chemical Process
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200241
| |
Collapse
|
9
|
Siddiqui AJ, Adnan M, Jahan S, Redman W, Saeed M, Patel M. Neurological disorder and psychosocial aspects of cerebral malaria: what is new on its pathogenesis and complications? A minireview. Folia Parasitol (Praha) 2020; 67. [PMID: 32636351 DOI: 10.14411/fp.2020.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/06/2020] [Indexed: 01/21/2023]
Abstract
Recently, malaria is remain considered as the most prevalent infectious disease, affecting the human health globally. High morbidity and mortality worldwide is often allied with cerebral malaria (CM) based disorders of the central nervous system, especially across many tropical and sub-tropical regions. These disorders are characterised by the infection of Plasmodium species, which leads to acute or chronic neurological disorders, even after having active/effective antimalarial drugs. Furthermore, even during the treatment, individual remain sensitive for neurological impairments in the form of decrease blood flow and vascular obstruction in brain including many more other changes. This review briefly explains and update on the epidemiology, burden of disease, pathogenesis and role of CM in neurological disorders with behaviour and function in mouse and human models. Moreover, the social stigma, which plays an important role in neurological disorders and a factor for assessing CM, is also discussed in this review.
Collapse
Affiliation(s)
| | | | - Sadaf Jahan
- Department of Medical Laboratory, College of Applied Medical Sciences, Majmaah University, Majmaah city, Saudi Arabia
| | - Whitni Redman
- Surgery Department, Division of Biomedical Research, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Mοhd Saeed
- Department of Biology, College of Science, University of Hail, Hail, PO Box 2440, Saudi Arabia
| | - Mitesh Patel
- Bapalal Vaidya Botanical Research Centre, Department of Biosciences, Veer Narmad South Gujarat University, Surat, Gujarat, India
| |
Collapse
|
10
|
Yadav DK, Kumar S, Teli MK, Yadav R, Chaudhary S. Molecular Targets for Malarial Chemotherapy: A Review. Curr Top Med Chem 2019; 19:861-873. [DOI: 10.2174/1568026619666190603080000] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 11/22/2022]
Abstract
The malaria parasite resistance to the existing drugs is a serious problem to the currently used
antimalarials and, thus, highlights the urgent need to develop new and effective anti-malarial molecules.
This could be achieved either by the identification of the new drugs for the validated targets or by further
refining/improving the existing antimalarials; or by combining previously effective agents with
new/existing drugs to have a synergistic effect that counters parasite resistance; or by identifying novel
targets for the malarial chemotherapy. In this review article, a comprehensive collection of some of the
novel molecular targets has been enlisted for the antimalarial drugs. The targets which could be deliberated
for developing new anti-malarial drugs could be: membrane biosynthesis, mitochondrial system,
apicoplasts, parasite transporters, shikimate pathway, hematin crystals, parasite proteases, glycolysis,
isoprenoid synthesis, cell cycle control/cycline dependent kinase, redox system, nucleic acid metabolism,
methionine cycle and the polyamines, folate metabolism, the helicases, erythrocyte G-protein, and
farnesyl transferases. Modern genomic tools approaches such as structural biology and combinatorial
chemistry, novel targets could be identified followed by drug development for drug resistant strains providing
wide ranges of novel targets in the development of new therapy. The new approaches and targets
mentioned in the manuscript provide a basis for the development of new unique strategies for antimalarial
therapy with limited off-target effects in the near future.
Collapse
Affiliation(s)
- Dharmendra K. Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Surendra Kumar
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Mahesh K. Teli
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Ravikant Yadav
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur-302017, India
| | - Sandeep Chaudhary
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur-302017, India
| |
Collapse
|
11
|
Kalaria PN, Karad SC, Raval DK. A review on diverse heterocyclic compounds as the privileged scaffolds in antimalarial drug discovery. Eur J Med Chem 2018; 158:917-936. [PMID: 30261467 DOI: 10.1016/j.ejmech.2018.08.040] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/18/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022]
Abstract
The upward extend of malaria collectively with the emergence of resistance against predictable drugs has put enormous pressure on public health systems to introduce new malaria treatments. Heterocycles play an important role in the design and discovery of new malaria active compounds. Heterocyclic compounds have attracted significant attention for malaria treatment because of simplicity of parallelization and the examining power with regard to chemical space. Introduction of a variety of heterocyclic compounds have enabled to maintain the high levels of antimalarial potency observed for other more lipophilic analogues whilst improving the solubility and the oral bioavailability in pre-clinical species. In this review, we present an overview of recent literature to provide imminent into the applications of different heterocyclic scaffolds in fighting against malaria.
Collapse
Affiliation(s)
- Piyush N Kalaria
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, 388120, Gujarat, India.
| | - Sharad C Karad
- Department of Chemistry, Marwadi University, Rajkot, Gujarat, India.
| | - Dipak K Raval
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, 388120, Gujarat, India.
| |
Collapse
|
12
|
Leveridge M, Chung CW, Gross JW, Phelps CB, Green D. Integration of Lead Discovery Tactics and the Evolution of the Lead Discovery Toolbox. SLAS DISCOVERY 2018; 23:881-897. [PMID: 29874524 DOI: 10.1177/2472555218778503] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There has been much debate around the success rates of various screening strategies to identify starting points for drug discovery. Although high-throughput target-based and phenotypic screening has been the focus of this debate, techniques such as fragment screening, virtual screening, and DNA-encoded library screening are also increasingly reported as a source of new chemical equity. Here, we provide examples in which integration of more than one screening approach has improved the campaign outcome and discuss how strengths and weaknesses of various methods can be used to build a complementary toolbox of approaches, giving researchers the greatest probability of successfully identifying leads. Among others, we highlight case studies for receptor-interacting serine/threonine-protein kinase 1 and the bromo- and extra-terminal domain family of bromodomains. In each example, the unique insight or chemistries individual approaches provided are described, emphasizing the synergy of information obtained from the various tactics employed and the particular question each tactic was employed to answer. We conclude with a short prospective discussing how screening strategies are evolving, what this screening toolbox might look like in the future, how to maximize success through integration of multiple tactics, and scenarios that drive selection of one combination of tactics over another.
Collapse
Affiliation(s)
- Melanie Leveridge
- 1 GlaxoSmithKline Drug Design and Selection, Platform Technology and Science, Stevenage, Hertfordshire, UK
| | - Chun-Wa Chung
- 1 GlaxoSmithKline Drug Design and Selection, Platform Technology and Science, Stevenage, Hertfordshire, UK
| | - Jeffrey W Gross
- 2 GlaxoSmithKline Drug Design and Selection, Platform Technology and Science, Collegeville, PA, USA
| | - Christopher B Phelps
- 3 GlaxoSmithKline Drug Design and Selection, Platform Technology and Science, Cambridge, MA, USA
| | - Darren Green
- 1 GlaxoSmithKline Drug Design and Selection, Platform Technology and Science, Stevenage, Hertfordshire, UK
| |
Collapse
|
13
|
Xue Y, Wang Y, Cao Z, Zhou J, Chen ZX. Computational insight into the cooperative role of non-covalent interactions in the aza-Henry reaction catalyzed by quinine derivatives: mechanism and enantioselectivity. Org Biomol Chem 2018; 14:9588-9597. [PMID: 27714327 DOI: 10.1039/c6ob01611a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Density functional theory (DFT) calculations were performed to elucidate the mechanism and the origin of the high enantioselectivity of the aza-Henry reaction of isatin-derived N-Boc ketimine catalyzed by a quinine-derived catalyst (QN). The C-C bond formation step is found to be both the rate-determining and the stereo-controlled step. The results revealed the important role of the phenolic OH group in pre-organizing the complex of nitromethane and QN and stabilizing the in situ-generated nitronate and protonated QN. Three possible activation modes for C-C bond formation involving different coordination patterns of catalyst and substrates were studied, and it was found that both the ion pair-hydrogen bonding mode and the Brønsted acid-hydrogen bonding mode are viable, with the latter slightly preferred for the real catalytic system. The calculated enantiomeric excess (ee) favouring the S enantiomer is in good agreement with the experimental result. The high reactivity and enantioselectivity can be ascribed to the cooperative role of the multiple non-covalent interactions, including classical and non-classical H bonding as well as anionπ interactions. These results also highlight the importance of the inclusion of dispersion correction for achieving a reasonable agreement between theory and experiment for the current reaction.
Collapse
Affiliation(s)
- Yunsheng Xue
- Institute of Theoretical and Computational Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China. and School of Pharmacy, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou, 221004, P. R. China
| | - Yuhui Wang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, 3663 N. Zhongshan Road, Shanghai, 200062, P. R. China
| | - Zhongyan Cao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, 3663 N. Zhongshan Road, Shanghai, 200062, P. R. China
| | - Jian Zhou
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, 3663 N. Zhongshan Road, Shanghai, 200062, P. R. China
| | - Zhao-Xu Chen
- Institute of Theoretical and Computational Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China.
| |
Collapse
|
14
|
Riniker S, Landrum GA, Montanari F, Villalba SD, Maier J, Jansen JM, Walters WP, Shelat AA. Virtual-screening workflow tutorials and prospective results from the Teach-Discover-Treat competition 2014 against malaria. F1000Res 2017; 6:1136. [PMID: 28928948 DOI: 10.12688/f1000research.11905.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2017] [Indexed: 12/21/2022] Open
Abstract
The first challenge in the 2014 competition launched by the Teach-Discover-Treat (TDT) initiative asked for the development of a tutorial for ligand-based virtual screening, based on data from a primary phenotypic high-throughput screen (HTS) against malaria. The resulting Workflows were applied to select compounds from a commercial database, and a subset of those were purchased and tested experimentally for anti-malaria activity. Here, we present the two most successful Workflows, both using machine-learning approaches, and report the results for the 114 compounds tested in the follow-up screen. Excluding the two known anti-malarials quinidine and amodiaquine and 31 compounds already present in the primary HTS, a high hit rate of 57% was found.
Collapse
Affiliation(s)
- Sereina Riniker
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | | | - Floriane Montanari
- Pharmacoinformatics Research Group, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Santiago D Villalba
- IMP - Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Julie Maier
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Johanna M Jansen
- Department of Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | | | - Anang A Shelat
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
15
|
Riniker S, Landrum GA, Montanari F, Villalba SD, Maier J, Jansen JM, Walters WP, Shelat AA. Virtual-screening workflow tutorials and prospective results from the Teach-Discover-Treat competition 2014 against malaria. F1000Res 2017. [PMID: 28928948 PMCID: PMC5580409 DOI: 10.12688/f1000research.11905.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The first challenge in the 2014 competition launched by the Teach-Discover-Treat (TDT) initiative asked for the development of a tutorial for ligand-based virtual screening, based on data from a primary phenotypic high-throughput screen (HTS) against malaria. The resulting Workflows were applied to select compounds from a commercial database, and a subset of those were purchased and tested experimentally for anti-malaria activity. Here, we present the two most successful Workflows, both using machine-learning approaches, and report the results for the 114 compounds tested in the follow-up screen. Excluding the two known anti-malarials quinidine and amodiaquine and 31 compounds already present in the primary HTS, a high hit rate of 57% was found.
Collapse
Affiliation(s)
- Sereina Riniker
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | | | - Floriane Montanari
- Pharmacoinformatics Research Group, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Santiago D Villalba
- IMP - Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Julie Maier
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Johanna M Jansen
- Department of Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | | | - Anang A Shelat
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
16
|
7-Chloro-4-aminoquinoline γ-hydroxy-γ-lactam derived-tetramates as a new family of antimalarial compounds. Bioorg Med Chem Lett 2016; 26:5308-5311. [DOI: 10.1016/j.bmcl.2016.09.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 11/21/2022]
|
17
|
Boss C, Aissaoui H, Amaral N, Bauer A, Bazire S, Binkert C, Brun R, Bürki C, Ciana CL, Corminboeuf O, Delahaye S, Dollinger C, Fischli C, Fischli W, Flock A, Frantz MC, Girault M, Grisostomi C, Friedli A, Heidmann B, Hinder C, Jacob G, Le Bihan A, Malrieu S, Mamzed S, Merot A, Meyer S, Peixoto S, Petit N, Siegrist R, Trollux J, Weller T, Wittlin S. Discovery and Characterization of ACT-451840: an Antimalarial Drug with a Novel Mechanism of Action. ChemMedChem 2016; 11:1995-2014. [DOI: 10.1002/cmdc.201600298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/04/2016] [Indexed: 11/09/2022]
|
18
|
O'Neill PM, Ward SA. Ein Chinolincarboxamid als Breitbandwirkstoff gegen Malaria: Angriff in drei parasitären Stadien. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201507264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
19
|
O'Neill PM, Ward SA. A Quinoline Carboxamide Antimalarial Drug Candidate Uniquely Targets Plasmodia at Three Stages of the Parasite Life Cycle. Angew Chem Int Ed Engl 2015; 54:13504-6. [PMID: 26426264 DOI: 10.1002/anie.201507264] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Paul M O'Neill
- Department of Chemistry, University of Liverpool, Liverpool, L69 7ZD (UK).
| | - Stephen A Ward
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA (UK)
| |
Collapse
|
20
|
Narrowing the translational research gap: what needs to be done? Future Med Chem 2015; 7:1843-6. [PMID: 26393393 DOI: 10.4155/fmc.15.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
21
|
Spillman NJ, Kirk K. The malaria parasite cation ATPase PfATP4 and its role in the mechanism of action of a new arsenal of antimalarial drugs. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2015; 5:149-62. [PMID: 26401486 PMCID: PMC4559606 DOI: 10.1016/j.ijpddr.2015.07.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/03/2015] [Accepted: 07/08/2015] [Indexed: 11/28/2022]
Abstract
The intraerythrocytic malaria parasite, Plasmodium falciparum, maintains a low cytosolic Na(+) concentration and the plasma membrane P-type cation translocating ATPase 'PfATP4' has been implicated as playing a key role in this process. PfATP4 has been the subject of significant attention in recent years as mutations in this protein confer resistance to a growing number of new antimalarial compounds, including the spiroindolones, the pyrazoles, the dihydroisoquinolones, and a number of the antimalarial agents in the Medicines for Malaria Venture's 'Malaria Box'. On exposure of parasites to these compounds there is a rapid disruption of cytosolic Na(+). Whether, and if so how, such chemically distinct compounds interact with PfATP4, and how such interactions lead to parasite death, is not yet clear. The fact that multiple different chemical classes have converged upon PfATP4 highlights its significance as a potential target for new generation antimalarial agents. A spiroindolone (KAE609, now known as cipargamin) has progressed through Phase I and IIa clinical trials with favourable results. In this review we consider the physiological role of PfATP4, summarise the current repertoire of antimalarial compounds for which PfATP4 is implicated in their mechanism of action, and provide an outlook on translation from target identification in the laboratory to patient treatment in the field.
Collapse
Affiliation(s)
- Natalie Jane Spillman
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia ; Department of Medicine (Infectious Diseases), Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kiaran Kirk
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| |
Collapse
|
22
|
Perspective: Challenges and opportunities in TB drug discovery from phenotypic screening. Bioorg Med Chem 2014; 23:5087-97. [PMID: 25577708 DOI: 10.1016/j.bmc.2014.12.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 02/06/2023]
Abstract
Tuberculosis poses a major global health problem and multi-drug resistant strains are increasingly prevalent. Hence there is an urgent need to discover new TB drugs. Cell based phenotypic screening represents a powerful approach to identify anti-mycobacterial compounds and elucidate novel targets. Three high throughput phenotypic screens were performed at NITD against mycobacterium. Hits were identified and chemical series selected for optimisation. This produced compounds with good in vitro anti-mycobacterial activity and pharmacokinetic properties. Some compounds displayed oral activity in mouse efficacy models of TB. Herein, we review the TB discovery efforts at NITD and share experiences in optimisation of phenotypic hits, describing challenges encountered and lessons learned. We also offer perspectives to facilitate future selection and advancement of phenotypic hits.
Collapse
|
23
|
(+)-SJ733, a clinical candidate for malaria that acts through ATP4 to induce rapid host-mediated clearance of Plasmodium. Proc Natl Acad Sci U S A 2014; 111:E5455-62. [PMID: 25453091 DOI: 10.1073/pnas.1414221111] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Drug discovery for malaria has been transformed in the last 5 years by the discovery of many new lead compounds identified by phenotypic screening. The process of developing these compounds as drug leads and studying the cellular responses they induce is revealing new targets that regulate key processes in the Plasmodium parasites that cause malaria. We disclose herein that the clinical candidate (+)-SJ733 acts upon one of these targets, ATP4. ATP4 is thought to be a cation-transporting ATPase responsible for maintaining low intracellular Na(+) levels in the parasite. Treatment of parasitized erythrocytes with (+)-SJ733 in vitro caused a rapid perturbation of Na(+) homeostasis in the parasite. This perturbation was followed by profound physical changes in the infected cells, including increased membrane rigidity and externalization of phosphatidylserine, consistent with eryptosis (erythrocyte suicide) or senescence. These changes are proposed to underpin the rapid (+)-SJ733-induced clearance of parasites seen in vivo. Plasmodium falciparum ATPase 4 (pfatp4) mutations that confer resistance to (+)-SJ733 carry a high fitness cost. The speed with which (+)-SJ733 kills parasites and the high fitness cost associated with resistance-conferring mutations appear to slow and suppress the selection of highly drug-resistant mutants in vivo. Together, our data suggest that inhibitors of PfATP4 have highly attractive features for fast-acting antimalarials to be used in the global eradication campaign.
Collapse
|
24
|
Westermaier Y, Barril X, Scapozza L. Virtual screening: an in silico tool for interlacing the chemical universe with the proteome. Methods 2014; 71:44-57. [PMID: 25193260 DOI: 10.1016/j.ymeth.2014.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/16/2014] [Accepted: 08/02/2014] [Indexed: 12/28/2022] Open
Abstract
In silico screening both in the forward (traditional virtual screening) and reverse sense (inverse virtual screening (IVS)) are helpful techniques for interlacing the chemical universe of small molecules with the proteome. The former, which is using a protein structure and a large chemical database, is well-known by the scientific community. We have chosen here to provide an overview on the latter, focusing on validation and target prioritization strategies. By comparing it to complementary or alternative wet-lab approaches, we put IVS in the broader context of chemical genomics, target discovery and drug design. By giving examples from the literature and an own example on how to validate the approach, we provide guidance on the issues related to IVS.
Collapse
Affiliation(s)
- Yvonne Westermaier
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva 4, Switzerland; Computational Biology & Drug Design Group, Departament de Fisicoquímica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.
| | - Xavier Barril
- Computational Biology & Drug Design Group, Departament de Fisicoquímica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva 4, Switzerland.
| |
Collapse
|
25
|
Abstract
Analysis of the origins of new drugs approved by the US Food and Drug Administration (FDA) from 1999 to 2008 suggested that phenotypic screening strategies had been more productive than target-based approaches in the discovery of first-in-class small-molecule drugs. However, given the relatively recent introduction of target-based approaches in the context of the long time frames of drug development, their full impact might not yet have become apparent. Here, we present an analysis of the origins of all 113 first-in-class drugs approved by the FDA from 1999 to 2013, which shows that the majority (78) were discovered through target-based approaches (45 small-molecule drugs and 33 biologics). In addition, of 33 drugs identified in the absence of a target hypothesis, 25 were found through a chemocentric approach in which compounds with known pharmacology served as the starting point, with only eight coming from what we define here as phenotypic screening: testing a large number of compounds in a target-agnostic assay that monitors phenotypic changes. We also discuss the implications for drug discovery strategies, including viewing phenotypic screening as a novel discipline rather than as a neoclassical approach.
Collapse
|
26
|
Njoroge M, Njuguna NM, Mutai P, Ongarora DSB, Smith PW, Chibale K. Recent approaches to chemical discovery and development against malaria and the neglected tropical diseases human African trypanosomiasis and schistosomiasis. Chem Rev 2014; 114:11138-63. [PMID: 25014712 DOI: 10.1021/cr500098f] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | - Paul W Smith
- Novartis Institute for Tropical Diseases , Singapore 138670, Singapore
| | | |
Collapse
|
27
|
Selection and optimization of hits from a high-throughput phenotypic screen against Trypanosoma cruzi. Future Med Chem 2014; 5:1733-52. [PMID: 24144410 DOI: 10.4155/fmc.13.139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Inhibitors of Trypanosoma cruzi with novel mechanisms of action are urgently required to diversify the current clinical and preclinical pipelines. Increasing the number and diversity of hits available for assessment at the beginning of the discovery process will help to achieve this aim. RESULTS We report the evaluation of multiple hits generated from a high-throughput screen to identify inhibitors of T. cruzi and from these studies the discovery of two novel series currently in lead optimization. Lead compounds from these series potently and selectively inhibit growth of T. cruzi in vitro and the most advanced compound is orally active in a subchronic mouse model of T. cruzi infection. CONCLUSION High-throughput screening of novel compound collections has an important role to play in diversifying the trypanosomatid drug discovery portfolio. A new T. cruzi inhibitor series with good drug-like properties and promising in vivo efficacy has been identified through this process.
Collapse
|
28
|
Duardo-Sánchez A, Munteanu CR, Riera-Fernández P, López-Díaz A, Pazos A, González-Díaz H. Modeling Complex Metabolic Reactions, Ecological Systems, and Financial and Legal Networks with MIANN Models Based on Markov-Wiener Node Descriptors. J Chem Inf Model 2013; 54:16-29. [DOI: 10.1021/ci400280n] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Aliuska Duardo-Sánchez
- Department
of Information and Communication Technologies, Computer Science Faculty, University of A Coruña, Campus de Elviña, 15071, A Coruña, A Coruña, Spain
- Department of Special Public Law, Financial
and Tributary Law Area, Faculty of Law, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, A Coruña, Spain
| | - Cristian R. Munteanu
- Department
of Information and Communication Technologies, Computer Science Faculty, University of A Coruña, Campus de Elviña, 15071, A Coruña, A Coruña, Spain
| | - Pablo Riera-Fernández
- Department
of Information and Communication Technologies, Computer Science Faculty, University of A Coruña, Campus de Elviña, 15071, A Coruña, A Coruña, Spain
| | - Antonio López-Díaz
- Department of Special Public Law, Financial
and Tributary Law Area, Faculty of Law, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, A Coruña, Spain
| | - Alejandro Pazos
- Department
of Information and Communication Technologies, Computer Science Faculty, University of A Coruña, Campus de Elviña, 15071, A Coruña, A Coruña, Spain
| | - Humberto González-Díaz
- Department of Organic Chemistry II, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Bizkaia, Spain
- IKERBASQUE, Basque
Foundation for Science, 48011, Bilbao, Biscay, Spain
| |
Collapse
|
29
|
Kanishchev OS, Lavoignat A, Picot S, Médebielle M, Bouillon JP. New route to the 5-((arylthio- and heteroarylthio)methylene)-3-(2,2,2-trifluoroethyl)-furan-2(5H)-ones—Key intermediates in the synthesis of 4-aminoquinoline γ-lactams as potent antimalarial compounds. Bioorg Med Chem Lett 2013; 23:6167-71. [DOI: 10.1016/j.bmcl.2013.08.108] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/27/2013] [Accepted: 08/29/2013] [Indexed: 11/24/2022]
|
30
|
Spitzmüller A, Mestres J. Prediction of the P. falciparum target space relevant to malaria drug discovery. PLoS Comput Biol 2013; 9:e1003257. [PMID: 24146604 PMCID: PMC3798273 DOI: 10.1371/journal.pcbi.1003257] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/20/2013] [Indexed: 11/18/2022] Open
Abstract
Malaria is still one of the most devastating infectious diseases, affecting hundreds of millions of patients worldwide. Even though there are several established drugs in clinical use for malaria treatment, there is an urgent need for new drugs acting through novel mechanisms of action due to the rapid development of resistance. Resistance emerges when the parasite manages to mutate the sequence of the drug targets to the extent that the protein can still perform its function in the parasite but can no longer be inhibited by the drug, which then becomes almost ineffective. The design of a new generation of malaria drugs targeting multiple essential proteins would make it more difficult for the parasite to develop full resistance without lethally disrupting some of its vital functions. The challenge is then to identify which set of Plasmodium falciparum proteins, among the millions of possible combinations, can be targeted at the same time by a given chemotype. To do that, we predicted first the targets of the close to 20,000 antimalarial hits identified recently in three independent phenotypic screening campaigns. All targets predicted were then projected onto the genome of P. falciparum using orthologous relationships. A total of 226 P. falciparum proteins were predicted to be hit by at least one compound, of which 39 were found to be significantly enriched by the presence and degree of affinity of phenotypically active compounds. The analysis of the chemically compatible target combinations containing at least one of those 39 targets led to the identification of a priority set of 64 multi-target profiles that can set the ground for a new generation of more robust malaria drugs.
Collapse
Affiliation(s)
- Andreas Spitzmüller
- Chemotargets SL and Systems Pharmacology, Research Programme on Biomedical Informatics (GRIB), IMIM Hospital del Mar Research Institute and Universitat Pompeu Fabra, Parc de Recerca Biomèdica, Barcelona, Catalonia, Spain
| | - Jordi Mestres
- Chemotargets SL and Systems Pharmacology, Research Programme on Biomedical Informatics (GRIB), IMIM Hospital del Mar Research Institute and Universitat Pompeu Fabra, Parc de Recerca Biomèdica, Barcelona, Catalonia, Spain
- * E-mail:
| |
Collapse
|
31
|
Teasdale ME, Prudhomme J, Torres M, Braley M, Cervantes S, Bhatia SC, La Clair JJ, Le Roch K, Kubanek J. Pharmacokinetics, metabolism, and in vivo efficacy of the antimalarial natural product bromophycolide A. ACS Med Chem Lett 2013; 4:989-993. [PMID: 24159368 DOI: 10.1021/ml4002858] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A suite of pharmacokinetic and pharmacological studies show that bromophycolide A (1), an inhibitor of drug-sensitive and drug-resistant Plasmodium falciparum, displays a typical small molecule profile with low toxicity and good bioavailability. Despite susceptibility to liver metabolism and a short in vivo half-life, 1 significantly decreased parasitemia in a malaria mouse model. Combining these data with prior SAR analyses, we demonstrate the potential for future development of 1 and its bioactive ester analogs.
Collapse
Affiliation(s)
- Margaret E. Teasdale
- School of Biology and School of Chemistry
and Biochemistry, Aquatic Chemical Ecology Center and
Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Jacques Prudhomme
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521, United States
| | - Manuel Torres
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521, United States
| | - Matthew Braley
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521, United States
| | - Serena Cervantes
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521, United States
| | - Shanti C. Bhatia
- School of Biology and School of Chemistry
and Biochemistry, Aquatic Chemical Ecology Center and
Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - James J. La Clair
- Xenobe Research Institute, San Diego, California 92163, United States
| | - Karine Le Roch
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521, United States
| | - Julia Kubanek
- School of Biology and School of Chemistry
and Biochemistry, Aquatic Chemical Ecology Center and
Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
32
|
Butera JA. Phenotypic Screening as a Strategic Component of Drug Discovery Programs Targeting Novel Antiparasitic and Antimycobacterial Agents: An Editorial. J Med Chem 2013; 56:7715-8. [DOI: 10.1021/jm400443k] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- John A. Butera
- Hager Biosciences, LLC, 116 Research Drive, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
33
|
Abstract
The emergence of resistance to artemisinins and the renewed efforts to eradicate malaria demand the urgent development of new drugs. In this endeavour, the evaluation of efficacy in animal models is often a go/no go decision assay in drug discovery. This important role relies on the capability of animal models to assess the disposition, toxicology and efficacy of drugs in a single test. Although the relative merits of each efficacy model of malaria as human surrogate have been extensively discussed, there are no critical analyses on the use of such models in current drug discovery. In this article, we intend to analyse how efficacy models are used to discover new antimalarial drugs. Our analysis indicates that testing drug efficacy is often the last assay in each discovery stage and the experimental designs utilized are not optimized to expedite decision-making and inform clinical development. In light of this analysis, we propose new ways to accelerate drug discovery using efficacy models.
Collapse
|
34
|
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
|
35
|
Abstract
There is an urgent need for the development of new antimalarial drugs with novel modes of actions. The malarial parasite, Plasmodium falciparum, has a relatively small kinome of <100 kinases, with many members exhibiting a high degree of structural divergence from their host counterparts. A number of Plasmodium kinases have recently been shown by reverse genetics to be essential for various parts of the complex parasitic life cycle, and are thus genetically validated as potential targets. Implementation of mass spectrometry-based phosphoproteomics approaches has informed on key phospho-signalling pathways in the parasite. In addition, global phenotypic screens have revealed a large number of putative protein kinase inhibitors with antimalarial potency. Taken together, these investigations point to the Plasmodium kinome as a rich source of potential new targets. In this review, we highlight recent progress made towards this goal.
Collapse
|
36
|
David-Bosne S, Florent I, Lund-Winther AM, Hansen JB, Buch-Pedersen M, Machillot P, le Maire M, Jaxel C. Antimalarial screening via large-scale purification of Plasmodium falciparum Ca2+-ATPase 6 and in vitro studies. FEBS J 2013; 280:5419-29. [PMID: 23497141 DOI: 10.1111/febs.12244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 02/18/2013] [Accepted: 03/11/2013] [Indexed: 02/04/2023]
Abstract
The most severe form of human malaria is caused by the parasite Plasmodium falciparum. Despite the current need, there is no effective vaccine and parasites are becoming resistant to most of the antimalarials available. Therefore, there is an urgent need to discover new drugs from targets that have not yet suffered from drug pressure with the aim of overcoming the problem of new emerging resistance. Membrane transporters, such as P. falciparum Ca(2+)-ATPase 6 (PfATP6), the P. falciparum sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase (SERCA), have been proposed as potentially good antimalarial targets. The present investigation focuses on: (a) the large-scale purification of PfATP6 for maintenance of its enzymatic activity; (b) screening for PfATP6 inhibitors from a compound library; and (c) the selection of the best inhibitors for further tests on P. falciparum growth in vitro. We managed to heterologously express in yeast and purify an active form of PfATP6 as previously described, although in larger amounts. In addition to some classical SERCA inhibitors, a chemical library of 1680 molecules was screened. From these, we selected a pool of the 20 most potent inhibitors of PfATP6, presenting half maximal inhibitory concentration values in the range 1-9 μm. From these, eight were chosen for evaluation of their effect on P. falciparum growth in vitro, and the best compound presented a half maximal inhibitory concentration of ~ 2 μm. We verified the absence of an inhibitory effect of most of the compounds on mammalian SERCA1a, representing a potential advantage in terms of human toxicity. The present study describes a multidisciplinary approach allowing the selection of promising PfATP6-specific inhibitors with good antimalarial activity.
Collapse
|
37
|
Calderón F, Wilson DM, Gamo FJ. Antimalarial drug discovery: recent progress and future directions. PROGRESS IN MEDICINAL CHEMISTRY 2013; 52:97-151. [PMID: 23384667 DOI: 10.1016/b978-0-444-62652-3.00003-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Félix Calderón
- Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Spain
| | | | | |
Collapse
|
38
|
Antimalarial aminothiazoles and aminopyridines from phenotypic whole-cell screening of a SoftFocus® library. Future Med Chem 2012; 4:2265-77. [DOI: 10.4155/fmc.12.176] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The current state of antimalarial drug resistance emphasizes the need for new therapies with novel modes of action that will add a significant benefit compared with current standards. In this regard, high throughput phenotypic whole-cell screening aids the discovery of novel antiplasmodial scaffolds that are inherently suited to hit-to-lead and lead-optimization efforts. The aminothiazoles and aminopyridines exemplify two such compound classes stemming from whole-cell screening. Respective structure–activity relationship determinations and subsequent optimization around these scaffolds led to frontrunner compounds in each series, which possess the desired antimalarial efficacy, bioavailability and metabolic stability to further progress medicinal chemistry programs.
Collapse
|
39
|
Phillips MA. Stoking the drug target pipeline for human African trypanosomiasis. Mol Microbiol 2012; 86:10-4. [PMID: 22925123 DOI: 10.1111/mmi.12001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2012] [Indexed: 11/28/2022]
Abstract
Trypanosoma brucei is the causative agent of African sleeping sickness, putting at risk up to 50 million people in sub-Saharan Africa. Current drug therapies are limited by toxicity and difficult treatment regimes and as the development of vaccines remains unlikely, the identification of better drugs to control this deadly disease is needed. Strategies for the identification of new lead compounds include phenotypic screening or target-based approaches. Implementation of the latter has been hampered by the lack of defined targets that are both essential and druggable. In this issue of Molecular Microbiology, Jones et al. (2012) report on the characterization of T. brucei pyridoxal kinase (PdxK), an enzyme required for the salvage of vitamin B6, an essential enzymatic cofactor. Genetic knock-down and small molecule inhibitor studies were used to demonstrate that PdxK is essential for parasite growth both in vitro and in a mouse model, providing both genetic and chemical validation of the target. An enzyme assay compatible with high-throughput screening (HTS) was developed and the X-ray crystal structure solved, showing the potential for species selective inhibition. These studies add a greatly needed additional target into the drug discovery pipeline for this deadly parasitic infection.
Collapse
Affiliation(s)
- Margaret A Phillips
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041, USA.
| |
Collapse
|