1
|
Luo AP, Giannangelo C, Siddiqui G, Creek DJ. Promising antimalarial hits from phenotypic screens: a review of recently-described multi-stage actives and their modes of action. Front Cell Infect Microbiol 2023; 13:1308193. [PMID: 38162576 PMCID: PMC10757594 DOI: 10.3389/fcimb.2023.1308193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Over the last two decades, global malaria cases caused by Plasmodium falciparum have declined due to the implementation of effective treatments and the use of insecticides. However, the COVID-19 pandemic caused major disruption in the timely delivery of medical goods and diverted public health resources, impairing malaria control. The emergence of resistance to all existing frontline antimalarials underpins an urgent need for new antimalarials with novel mechanisms of action. Furthermore, the need to reduce malaria transmission and/or prevent malaria infection has shifted the focus of antimalarial research towards the discovery of compounds that act beyond the symptomatic blood stage and also impact other parasite life cycle stages. Phenotypic screening has been responsible for the majority of new antimalarial lead compounds discovered over the past 10 years. This review describes recently reported novel antimalarial hits that target multiple parasite stages and were discovered by phenotypic screening during the COVID-19 pandemic. Their modes of action and targets in blood stage parasites are also discussed.
Collapse
Affiliation(s)
| | | | - Ghizal Siddiqui
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Darren J. Creek
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
2
|
Mandal A, Kushwaha R, Mandal AA, Bajpai S, Yadav AK, Banerjee S. Transition Metal Complexes as Antimalarial Agents: A Review. ChemMedChem 2023; 18:e202300326. [PMID: 37436090 DOI: 10.1002/cmdc.202300326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/13/2023]
Abstract
In antimalarial drug development research, overcoming drug resistance has been a major challenge for researchers. Nowadays, several drugs like chloroquine, mefloquine, sulfadoxine, and artemisinin are used to treat malaria. But increment in drug resistance has pushed researchers to find novel drugs to tackle drug resistance problems. The idea of using transition metal complexes with pharmacophores as ligands/ligand pendants to show enhanced antimalarial activity with a novel mechanism of action has gained significant attention recently. The advantages of metal complexes include tunable chemical/physical properties, redox activity, avoiding resistance factors, etc. Several recent reports have successfully demonstrated that the metal complexation of known organic antimalarial drugs can overcome drug resistance by showing enhanced activities than the parent drugs. This review has discussed the fruitful research works done in the past few years falling into this criterion. Based on transition metal series (3d, 4d, or 5d), the antimalarial metal complexes have been divided into three broad categories (3d, 4d, or 5d metal-based), and their activities have been compared with the similar control complexes as well as the parent drugs. Furthermore, we have also commented on the potential issues and their possible solution for translating these metal-based antimalarial complexes into the clinic.
Collapse
Affiliation(s)
- Apurba Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), 221005, Varanasi, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), 221005, Varanasi, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), 221005, Varanasi, India
| | - Sumit Bajpai
- Department of Chemistry, Indian Institute of Technology (BHU), 221005, Varanasi, India
| | - Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), 221005, Varanasi, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), 221005, Varanasi, India
| |
Collapse
|
3
|
Veale CGL, Olomola TO, Chellan P, Edkins AL. Biological and Medicinal Chemistry in Africa. Chembiochem 2023; 24:e202300060. [PMID: 36942876 DOI: 10.1002/cbic.202300060] [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: 02/02/2023] [Revised: 02/15/2023] [Indexed: 03/23/2023]
Abstract
The young, fast-growing population of Africa means that harnessing the economic benefits of scientific research is critical to sustained and equitable growth in the continent. Moreover, the whole world would benefit from the added intellectual contribution that would come from nurturing African science. The high burden of neglected diseases in Africa makes chemical biology a particularly important field. In this editorial, the reconvergence of science conducted at the interface of chemistry and biology is placed in the context of African participation, its importance to global science and the unique blend of supporting and hindering factors that influence African scientific contributions. The new Biological and Medicinal Chemistry in Africa special collection showcases a broad spectrum of African chemical biology.
Collapse
Affiliation(s)
- Clinton G L Veale
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Temitope O Olomola
- Department of Chemistry, Faculty of Science, Obafemi Awolowo University, Ile-Ile, 220005, Nigeria
| | - Prinessa Chellan
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, Stellenbosch, South Africa
| | - Adrienne L Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makanda, 6139, South Africa
| |
Collapse
|
4
|
Escala N, Pineda LM, Ng MG, Coronado LM, Spadafora C, del Olmo E. Antiplasmodial activity, structure-activity relationship and studies on the action of novel benzimidazole derivatives. Sci Rep 2023; 13:285. [PMID: 36609676 PMCID: PMC9822940 DOI: 10.1038/s41598-022-27351-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/30/2022] [Indexed: 01/08/2023] Open
Abstract
Malaria cases and deaths keep being excessively high every year. Some inroads gained in the last two decades have been eroded especially due to the surge of resistance to most antimalarials. The search for new molecules that can replace the ones currently in use cannot stop. In this report, the synthesis of benzimidazole derivatives guided by structure-activity parameters is presented. Thirty-six molecules obtained are analyzed according to their activity against P. falciparum HB3 strain based on the type of substituent on rings A and B, their electron donor/withdrawing, as well as their dimension/spatial properties. There is a preference for electron donating groups on ring A, such as Me in position 5, or better, 5, 6-diMe. Ring B must be of the pyridine type such as picolinamide, other modifications are generally not favorable. Two molecules, 1 and 33 displayed antiplasmodial activity in the high nanomolar range against the chloroquine sensitive strain, with selectivity indexes above 10. Activity results of 1, 12 and 16 on a chloroquine resistance strain indicated an activity close to chloroquine for compound 1. Analysis of some of their effect on the parasites seem to suggest that 1 and 33 affect only the parasite and use a route other than interference with hemozoin biocrystallization, the route used by chloroquine and most antimalarials.
Collapse
Affiliation(s)
- Nerea Escala
- grid.452531.4Departamento de Ciencias Farmacéuticas: Química Farmacéutica, Facultad de Farmacia, Universidad de Salamanca, CIETUS, IBSAL, 37007 Salamanca, Spain
| | - Laura M. Pineda
- Center of Cellular and Molecular Biology of Diseases, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología, City of Knowledge, Clayton, Apartado 0816-02852, Panama City, Panama
| | - Michelle G. Ng
- Center of Cellular and Molecular Biology of Diseases, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología, City of Knowledge, Clayton, Apartado 0816-02852, Panama City, Panama
| | - Lorena M. Coronado
- Center of Cellular and Molecular Biology of Diseases, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología, City of Knowledge, Clayton, Apartado 0816-02852, Panama City, Panama
| | - Carmenza Spadafora
- Center of Cellular and Molecular Biology of Diseases, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología, City of Knowledge, Clayton, Apartado 0816-02852, Panama City, Panama.
| | - Esther del Olmo
- grid.452531.4Departamento de Ciencias Farmacéuticas: Química Farmacéutica, Facultad de Farmacia, Universidad de Salamanca, CIETUS, IBSAL, 37007 Salamanca, Spain
| |
Collapse
|
5
|
Sousa CC, Dziwornu GA, Quadros HC, Araujo-Neto JH, Chibale K, Moreira DRM. Antimalarial Pyrido[1,2- a]benzimidazoles Exert Strong Parasiticidal Effects by Achieving High Cellular Uptake and Suppressing Heme Detoxification. ACS Infect Dis 2022; 8:1700-1710. [PMID: 35848708 DOI: 10.1021/acsinfecdis.2c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pyrido[1,2-a]benzimidazoles (PBIs) are synthetic antiplasmodium agents with potent activity and are structurally differentiated from benchmark antimalarials. To study the cellular uptake of PBIs and understand the underlying phenotype of their antiplasmodium activity, their antiparasitic activities were examined in chloroquine (CQ)-susceptible and CQ-resistant Plasmodium falciparum in vitro. Moreover, drug uptake and heme detoxification suppression were examined in Plasmodium berghei-infected mice. The in vitro potency of PBIs is comparable to most 4-aminoquinolines. They have a speed of action in vitro that is superior to that of atovaquone and an ability to kill rings and trophozoites. The antiparasitic effects observed for the PBIs in cell culture and in infected mice are similar in terms of potency and efficacy and are comparable to CQ but with the added advantage of demonstrating equipotency against both CQ susceptible and resistant parasite strains. PBIs have a high rate of uptake by parasite cells and, conversely, a limited rate of uptake by host cells. The mechanism of cellular uptake of the PBIs differs from the ion-trap mechanism typically observed for 4-aminoquinolines, although they share key structural features. The high cellular uptake, attractive parasiticidal profile, and susceptibility of resistant strains to PBIs are desirable characteristics for new antimalarial agents.
Collapse
Affiliation(s)
- Caroline C Sousa
- Fundação Oswaldo Cruz (Fiocruz), Instituto Gonçalo Moniz, Salvador, 40296-710 Bahia, Brazil
| | - Godwin Akpeko Dziwornu
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Helenita C Quadros
- Fundação Oswaldo Cruz (Fiocruz), Instituto Gonçalo Moniz, Salvador, 40296-710 Bahia, Brazil
| | | | - Kelly Chibale
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Diogo R M Moreira
- Fundação Oswaldo Cruz (Fiocruz), Instituto Gonçalo Moniz, Salvador, 40296-710 Bahia, Brazil
| |
Collapse
|
6
|
Bernard MM, Mohanty A, Rajendran V. Title: A Comprehensive Review on Classifying Fast-acting and Slow-acting Antimalarial Agents Based on Time of Action and Target Organelle of Plasmodium sp. Pathog Dis 2022; 80:6589403. [PMID: 35588061 DOI: 10.1093/femspd/ftac015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/20/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical resistance towards malarial parasites has rendered many antimalarials ineffective, likely due to a lack of understanding of time of action and stage specificity of all life stages. Therefore, to tackle this problem a more incisive comprehensive analysis of the fast and slow-acting profile of antimalarial agents relating to parasite time-kill kinetics and the target organelle on the progression of blood-stage parasites was carried out. It is evident from numerous findings that drugs targeting food vacuole, nuclear components, and endoplasmic reticulum mainly exhibit a fast-killing phenotype within 24h affecting first-cycle activity. Whereas drugs targeting mitochondria, apicoplast, microtubules, parasite invasion and egress exhibit a largely slow-killing phenotype within 96-120h, affecting second-cycle activity with few exemptions as moderately fast-killing. It is essential to understand the susceptibility of drugs on rings, trophozoites, schizonts, merozoites, and the appearance of organelle at each stage of 48h intraerythrocytic parasite cycle. Therefore, these parameters may facilitate the paradigm for understanding the timing of antimalarials action in deciphering its precise mechanism linked with time. Thus, classifying drugs based on the time of killing may promote designing new combination regimens against varied strains of P. falciparum and evaluating potential clinical resistance.
Collapse
Affiliation(s)
- Monika Marie Bernard
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Abhinab Mohanty
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Vinoth Rajendran
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| |
Collapse
|
7
|
Yun ES, Akhtar MS, Mohandoss S, Lee YR. Microwave-assisted annulation for the construction of pyrido-fused heterocycles and their application as photoluminescent chemosensors. Org Biomol Chem 2022; 20:3397-3407. [PMID: 35362508 DOI: 10.1039/d2ob00257d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A catalyst-free microwave-assisted annulation protocol for the preparation of biologically interesting pyrido-fused quinazolinones and pyrido[1,2-a]benzimidazoles is developed. This reaction involves the [3 + 3] annulation of various quinazolinones or benzimidazoles with 3-formylchromones to yield functionalized 11H-pyrido[2,1-b]quinazolin-11-one and pyrido[1,2-a] benzimidazole derivatives. This approach is successfully extended to the construction of various pyrazolo[4,3-d]pyrido[1,2-a]pyrimidin-10(1H)-ones. The present approach is complementary to the existing synthetic methodologies and offers a rapid and facile approach with a broad substrate scope, good yields, catalyst-free conditions, and a high functional group tolerance. The optimal synthesized compound is also employed as an "on-off" photoluminescent probe for the selective detection of Fe3+ and Ag+ metal ions.
Collapse
Affiliation(s)
- Ei Seul Yun
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Muhammad Saeed Akhtar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Sonaimuthu Mohandoss
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Yong Rok Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
8
|
Yang K, Chen ZX, Zhou YJ, Chen Q, Yu SW, Luo SH, Wang ZY. Simple inorganic base promoted polycyclic construction using mucohalic acid as a C 3 synthon: synthesis and AIE probe application of benzo[4,5]imidazo[1,2- a]pyridines. Org Chem Front 2022. [DOI: 10.1039/d1qo01753e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Using mucohalic acid as C3 synthon via a transition metal-free multicomponent reaction, an eco-friendly protocol to synthesize C1-functionalized benzo[4,5]imidazo[1,2-a]pyridines which can be applied as fluorescence probe for picric acid is described.
Collapse
Affiliation(s)
- Kai Yang
- School of Chemistry, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, Guangzhou 510006, P. R. China
- College of pharmacy, Gannan Medical University, Ganzhou 341000, P. R. China
| | - Zhi-Xi Chen
- College of pharmacy, Gannan Medical University, Ganzhou 341000, P. R. China
| | - Yong-Jun Zhou
- School of Chemistry, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, Guangzhou 510006, P. R. China
| | - Qi Chen
- School of Chemistry, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, Guangzhou 510006, P. R. China
| | - Shi-Wei Yu
- School of Chemistry, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, Guangzhou 510006, P. R. China
| | - Shi-He Luo
- School of Chemistry, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, Guangzhou 510006, P. R. China
| | - Zhao-Yang Wang
- School of Chemistry, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, Guangzhou 510006, P. R. China
| |
Collapse
|
9
|
Welsh A, Mbaba M, Prince S, Smith GS. Synthesis, molecular modeling and preliminary anticancer evaluation of 2-ferrocenylbenzimidazole metallofragments. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.131122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
10
|
Brishty SR, Hossain MJ, Khandaker MU, Faruque MRI, Osman H, Rahman SMA. A Comprehensive Account on Recent Progress in Pharmacological Activities of Benzimidazole Derivatives. Front Pharmacol 2021; 12:762807. [PMID: 34803707 PMCID: PMC8597275 DOI: 10.3389/fphar.2021.762807] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
Nowadays, nitrogenous heterocyclic molecules have attracted a great deal of interest among medicinal chemists. Among these potential heterocyclic drugs, benzimidazole scaffolds are considerably prevalent. Due to their isostructural pharmacophore of naturally occurring active biomolecules, benzimidazole derivatives have significant importance as chemotherapeutic agents in diverse clinical conditions. Researchers have synthesized plenty of benzimidazole derivatives in the last decades, amidst a large share of these compounds exerted excellent bioactivity against many ailments with outstanding bioavailability, safety, and stability profiles. In this comprehensive review, we have summarized the bioactivity of the benzimidazole derivatives reported in recent literature (2012-2021) with their available structure-activity relationship. Compounds bearing benzimidazole nucleus possess broad-spectrum pharmacological properties ranging from common antibacterial effects to the world's most virulent diseases. Several promising therapeutic candidates are undergoing human trials, and some of these are going to be approved for clinical use. However, notable challenges, such as drug resistance, costly and tedious synthetic methods, little structural information of receptors, lack of advanced software, and so on, are still viable to be overcome for further research.
Collapse
Affiliation(s)
- Shejuti Rahman Brishty
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Md. Jamal Hossain
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway, Malaysia
| | | | - Hamid Osman
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - S. M. Abdur Rahman
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
11
|
Leshabane M, Dziwornu GA, Coertzen D, Reader J, Moyo P, van der Watt M, Chisanga K, Nsanzubuhoro C, Ferger R, Erlank E, Venter N, Koekemoer L, Chibale K, Birkholtz LM. Benzimidazole Derivatives Are Potent against Multiple Life Cycle Stages of Plasmodium falciparum Malaria Parasites. ACS Infect Dis 2021; 7:1945-1955. [PMID: 33673735 DOI: 10.1021/acsinfecdis.0c00910] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The continued emergence of resistance to front-line antimalarial treatments is of great concern. Therefore, new compounds that potentially have a novel target in various developmental stages of Plasmodium parasites are needed to treat patients and halt the spread of malaria. Here, several benzimidazole derivatives were screened for activity against the symptom-causing intraerythrocytic asexual blood stages and the transmissible gametocyte stages of P. falciparum. Submicromolar activity was obtained for 54 compounds against asexual blood stage parasites with 6 potent at IC50 < 100 nM while not displaying any marked toxicity against mammalian cells. Nanomolar potency was also observed against gametocytes with two compounds active against early stage gametocytes and two compounds active against late-stage gametocytes. The transmission-blocking potential of the latter was confirmed as they could prevent male gamete exflagellation and the lead compound reduced transmission by 72% in an in vivo mosquito feeding model. These compounds therefore have activity against multiple stages of Plasmodium parasites with potential for differential targets.
Collapse
Affiliation(s)
- Meta Leshabane
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | | | - Dina Coertzen
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Phanankosi Moyo
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Kelly Chisanga
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Richard Ferger
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Erica Erlank
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, and Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Nelius Venter
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, and Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Lizette Koekemoer
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, and Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| |
Collapse
|
12
|
Probst A, Chisanga K, Dziwornu GA, Haeberli C, Keiser J, Chibale K. Expanding the Activity Profile of Pyrido[1,2- a]benzimidazoles: Synthesis and Evaluation of Novel N1-1-Phenylethanamine Derivatives against Schistosoma mansoni. ACS Infect Dis 2021; 7:1032-1043. [PMID: 32786285 DOI: 10.1021/acsinfecdis.0c00278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Praziquantel is the only widely available drug to treat schistosomiasis. With very few candidates currently in the drug development pipeline, there is an urgent need to discover and develop novel antischistosomal drugs. In this regard, the pyrido[1,2-a]benzimidazole (PBI) scaffold has emerged as a promising chemotype in hit-to-lead efforts. Here, we report a novel series of antischistosomal PBIs with potent in vitro activity (IC50 values of 0.08-1.43 μM) against Schistosoma mansoni newly transformed schistosomula and adult worms. Moreover, the current PBIs demonstrated good hepatic microsomal stability (>70% of drug remaining after 30 min) and were nontoxic to the Chinese hamster ovarian and human liver HepG2 cells, though toxicity (selectivity index, SI < 10) against the rat L6 myoblast cell line was observed. The compounds showed a small therapeutic window but were efficacious in vivo, exhibiting moderate to high worm burden reductions of 35.8-89.6% in S. mansoni-infected mice.
Collapse
Affiliation(s)
- Alexandra Probst
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, P.O. Box CH-4003, Basel, Switzerland
| | - Kelly Chisanga
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Cécile Haeberli
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, P.O. Box CH-4003, Basel, Switzerland
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, P.O. Box CH-4003, Basel, Switzerland
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery Unit, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
13
|
Dziwornu GA, Coertzen D, Leshabane M, Korkor CM, Cloete CK, Njoroge M, Gibhard L, Lawrence N, Reader J, van der Watt M, Wittlin S, Birkholtz LM, Chibale K. Antimalarial Benzimidazole Derivatives Incorporating Phenolic Mannich Base Side Chains Inhibit Microtubule and Hemozoin Formation: Structure-Activity Relationship and In Vivo Oral Efficacy Studies. J Med Chem 2021; 64:5198-5215. [PMID: 33844521 DOI: 10.1021/acs.jmedchem.1c00354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A novel series of antimalarial benzimidazole derivatives incorporating phenolic Mannich base side chains at the C2 position, which possess dual asexual blood and sexual stage activities, is presented. Structure-activity relationship studies revealed that the 1-benzylbenzimidazole analogues possessed submicromolar asexual blood and sexual stage activities in contrast to the 1H-benzimidazole analogues, which were only active against asexual blood stage (ABS) parasites. Further, the former demonstrated microtubule inhibitory activity in ABS parasites but more significantly in stage II/III gametocytes. In addition to being bona fide inhibitors of hemozoin formation, the 1H-benzimidazole analogues also showed inhibitory effects on microtubules. In vivo efficacy studies in Plasmodium berghei-infected mice revealed that the frontrunner compound 41 exhibited high efficacy (98% reduction in parasitemia) when dosed orally at 4 × 50 mg/kg. Generally, the compounds were noncytotoxic to mammalian cells.
Collapse
Affiliation(s)
| | - Dina Coertzen
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Meta Leshabane
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Constance M Korkor
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Cleavon K Cloete
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Liezl Gibhard
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Nina Lawrence
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, Basel 4002, Switzerland.,University of Basel, Basel 4003, Switzerland
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa.,South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
14
|
Alzain AA, Brisson L, Delaye PO, Pénichon M, Chadet S, Besson P, Chevalier S, Allouchi H, Mohamed MA, Roger S, Enguehard-Gueiffier C. Bioinspired imidazo[1,2-a:4,5-c']dipyridines with dual antiproliferative and anti-migrative properties in human cancer cells: The SAR investigation. Eur J Med Chem 2021; 218:113258. [PMID: 33813152 DOI: 10.1016/j.ejmech.2021.113258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/24/2022]
Abstract
Herein, we report the design, synthesis and evaluation of novel bioinspired imidazo[1,2-a:4,5c']dipyridines. The structural optimization identified four anti-proliferative compounds. Compounds 11, 18, 19 and 20 exhibited excellent anticancer activities in vitro with IC50 of 0.4-5 μM against three human cancer cell lines (MDA-MB-468, MDA-MB-435s and MDA-MB-231). These four compounds induced apoptosis in MDA-MB-231 cells in a dose-dependent manner, targeting different apoptotic proteins expression: 11 increased the expression of pro-apoptotic Bax protein while 18-20 reduced the level of anti-apoptotic Bcl-2 protein. Compounds 18 and 19 also reduced MDA-MB-231 cells proliferation as measured by Ki-67 staining. Furthermore, compounds were also tested for the ability to inhibit cell migration in the highly aggressive human MDA-MB-435s cell line. Six compounds of this series (8, 15, 18, 22, 23, 24) inhibited cell migration by 41-50% while four compounds (20, 25, 27, 30) inhibited the migration by 53-62% in wound-healing experiments. Interestingly, compound 20 presented both antiproliferative and anti-migration activities and might be a promising anti-metastatic agent for cancer treatment.
Collapse
Affiliation(s)
- Abdulrahim A Alzain
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France; University of Gezira, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, P.O box 20, Gezira, Sudan
| | - Lucie Brisson
- University of Tours, INSERM, UMR 1069 N2C, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Pierre-Olivier Delaye
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France
| | - Mélanie Pénichon
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France
| | - Stéphanie Chadet
- University of Tours, EA 4245 T2I, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Pierre Besson
- University of Tours, EA 4245 T2I, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Stéphan Chevalier
- University of Tours, INSERM, UMR 1069 N2C, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Hassan Allouchi
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France
| | - Magdi A Mohamed
- University of Khartoum, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Khartoum, Sudan; Jouf University, College of Pharmacy, Department of Pharmaceutical Chemistry, Saudi Arabia
| | - Sébastien Roger
- University of Tours, EA 4245 T2I, 10 boulevard Tonnellé, 37032, Tours Cedex, France; Institut Universitaire de France, 75006, Paris, France.
| | | |
Collapse
|
15
|
Korkor CM, Garnie LF, Amod L, Egan TJ, Chibale K. Intrinsic fluorescence properties of antimalarial pyrido[1,2- a]benzimidazoles facilitate subcellular accumulation and mechanistic studies in the human malaria parasite Plasmodium falciparum. Org Biomol Chem 2020; 18:8668-8676. [PMID: 33078179 PMCID: PMC7710849 DOI: 10.1039/d0ob01730b] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The intrinsic fluorescence properties of two related pyrido[1,2-a]benzimidazole antimalarial compounds suitable for the cellular imaging of the human malaria parasite Plasmodium falciparum without the need to attach extrinsic fluorophores are described. Although these compounds are structurally related, they have been shown by confocal microscopy to not only accumulate selectively within P. falciparum but to also accumulate differently in the organelles investigated. Localization to the digestive vacuole and nearby neutral lipids was observed for compound 2 which was shown to inhibit hemozoin formation using a cellular fractionation assay indicating that this is a contributing mechanism of action. By contrast, compound 1, which differs from compound 2 by the replacement of the imidazole[1,2-a:4,5-b']dipyridine core with the benzimidazole core as well as the presence of Cl substituents, shows very different localisation patterns and shows no evidence of hemozoin inhibition, suggesting a different mechanism of antimalarial action. Docking profiles of both compounds on the hemozoin surface further provided insight into their mechanisms of action.
Collapse
Affiliation(s)
- Constance M Korkor
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.
| | - Larnelle F Garnie
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.
| | - Leah Amod
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa. and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa. and South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Department of Chemistry, Rondebosch 7701, South Africa and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
16
|
Veale CGL, Müller R. Recent Highlights in Anti-infective Medicinal Chemistry from South Africa. ChemMedChem 2020; 15:809-826. [PMID: 32149446 DOI: 10.1002/cmdc.202000086] [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: 02/11/2020] [Indexed: 12/17/2022]
Abstract
Global advancements in biological technologies have vastly increased the variety of and accessibility to bioassay platforms, while simultaneously improving our understanding of druggable chemical space. In the South African context, this has resulted in a rapid expansion in the number of medicinal chemistry programmes currently operating, particularly on university campuses. Furthermore, the modern medicinal chemist has the advantage of being able to incorporate data from numerous related disciplines into the medicinal chemistry process, allowing for informed molecular design to play a far greater role than previously possible. Accordingly, this review focusses on recent highlights in drug-discovery programmes, in which South African medicinal chemistry groups have played a substantive role in the design and optimisation of biologically active compounds which contribute to the search for promising agents for infectious disease.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| | - Ronel Müller
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
17
|
Mueller R, Reddy V, Nchinda AT, Mebrahtu F, Taylor D, Lawrence N, Tanner L, Barnabe M, Eyermann CJ, Zou B, Kondreddi RR, Lakshminarayana SB, Rottmann M, Street LJ, Chibale K. Lerisetron Analogues with Antimalarial Properties: Synthesis, Structure-Activity Relationship Studies, and Biological Assessment. ACS OMEGA 2020; 5:6967-6982. [PMID: 32258933 PMCID: PMC7114883 DOI: 10.1021/acsomega.0c00327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/05/2020] [Indexed: 06/11/2023]
Abstract
A phenotypic whole cell high-throughput screen against the asexual blood and liver stages of the malaria parasite identified a benzimidazole chemical series. Among the hits were the antiemetic benzimidazole drug Lerisetron 1 (IC50 NF54 = 0.81 μM) and its methyl-substituted analogue 2 (IC50 NF54 = 0.098 μM). A medicinal chemistry hit to lead effort led to the identification of chloro-substituted analogue 3 with high potency against the drug-sensitive NF54 (IC50 NF54 = 0.062 μM) and multidrug-resistant K1 (IC50 K1 = 0.054 μM) strains of the human malaria parasite Plasmodium falciparum. Compounds 2 and 3 gratifyingly showed in vivo efficacy in both Plasmodium berghei and P. falciparum mouse models of malaria. Cardiotoxicity risk as expressed in strong inhibition of the human ether-a-go-go-related gene (hERG) potassium channel was identified as a major liability to address. This led to the synthesis and biological assessment of around 60 analogues from which several compounds with improved antiplasmodial potency, relative to the lead compound 3, were identified.
Collapse
Affiliation(s)
- Rudolf Mueller
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Virsinha Reddy
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Aloysius T. Nchinda
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Fanuel Mebrahtu
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Dale Taylor
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Nina Lawrence
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Lloyd Tanner
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Marine Barnabe
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Charles J. Eyermann
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Bin Zou
- Shanghai
Blueray Biopharma Co. LTD, Block 3, 576 Libing Road, Pudong New District, Shanghai 201301, China
| | - Ravinder R. Kondreddi
- PJS
Pharma Pvt. Ltd., Plot
No. 103/1, Phase II, IDA Cherlapally, Hyderabad 500051, India
| | - Suresh B. Lakshminarayana
- Novartis
Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Matthias Rottmann
- Department
of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4051 Basel, Switzerland
- University
of Basel, 4002 Basel, Switzerland
| | - Leslie J. Street
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South
African Medical Research Council Drug Discovery and Development Research
Unit, Department of Chemistry & Institute of Infectious Disease
and Molecular Medicine, University of Cape
Town, Rondebosch 7701, South Africa
| |
Collapse
|
18
|
Virtual screening as a tool to discover new β-haematin inhibitors with activity against malaria parasites. Sci Rep 2020; 10:3374. [PMID: 32099045 PMCID: PMC7042288 DOI: 10.1038/s41598-020-60221-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 02/10/2020] [Indexed: 12/24/2022] Open
Abstract
Malaria remains a major public health problem. With the loss of antimalarials to resistance, the malaria burden will likely continue for decades. New antimalarial scaffolds are crucial to avoid cross-resistance. Here, we present the first structure based virtual screening using the β-haematin crystal as a target for new inhibitor scaffolds by applying a docking method. The ZINC15 database was searched for compounds with high binding affinity with the surface of the β-haematin crystal using the PyRx Virtual Screening Tool. Top-ranked compounds predicted to interact with β-haematin were submitted to a second screen applying in silico toxicity and drug-likeness predictions using Osiris DataWarrior. Fifteen compounds were purchased for experimental testing. An NP-40 mediated β-haematin inhibition assay and parasite growth inhibition activity assay were performed. The benzoxazole moiety was found to be a promising scaffold for further development, showing intraparasitic haemozoin inhibition using a cellular haem fractionation assay causing a decrease in haemozoin in a dose dependent manner with a corresponding increase in exchangeable haem. A β-haematin inhibition hit rate of 73% was found, a large enrichment over random screening, demonstrating that virtual screening can be a useful and cost-effective approach in the search for new haemozoin inhibiting antimalarials.
Collapse
|
19
|
Sharma M, Prasher P. An epigrammatic status of the ' azole'-based antimalarial drugs. RSC Med Chem 2020; 11:184-211. [PMID: 33479627 PMCID: PMC7536834 DOI: 10.1039/c9md00479c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/26/2019] [Indexed: 11/21/2022] Open
Abstract
The development of multidrug resistance in the malarial parasite has sabotaged majority of the eradication efforts by restraining the inhibition profile of first line as well as second line antimalarial drugs, thus necessitating the development of novel pharmaceutics constructed on appropriate scaffolds with superior potency against the drug-resistant and drug-susceptible Plasmodium parasite. Over the past decades, the infectious malarial parasite has developed resistance against most of the contemporary therapeutics, thus necessitating the rational development of novel approaches principally focused on MDR malaria. This review presents an epigrammatic collation of the epidemiology and the contemporary antimalarial therapeutics based on the 'azole' motif.
Collapse
Affiliation(s)
- Mousmee Sharma
- Department of Chemistry , Uttaranchal University , Dehradun 248007 , India
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
| | - Parteek Prasher
- Department of Chemistry , University of Petroleum & Energy Studies , Dehradun 248007 , India . ;
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
| |
Collapse
|
20
|
Ni S, Li B, Xu Y, Mao F, Li X, Lan L, Zhu J, Li J. Targeting virulence factors as an antimicrobial approach: Pigment inhibitors. Med Res Rev 2019; 40:293-338. [PMID: 31267561 DOI: 10.1002/med.21621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/30/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022]
Abstract
The fascinating and dangerous colored pathogens contain unique chemically pigmented molecules, which give varied and efficient assistance as virulence factors to the crucial reproduction and growth of microbes. Therefore, multiple novel strategies and inhibitors have been developed in recent years that target virulence factor pigments. However, despite the importance and significance of this topic, it has not yet been comprehensively reviewed. Moreover, research groups around the world have made successful progress against antibacterial infections by targeting pigment production, including our serial works on the discovery of CrtN inhibitors against staphyloxanthin production in Staphylococcus aureus. On the basis of the previous achievements and recent progress of our group in this field, this article will be the first comprehensive review of pigment inhibitors against colored pathogens, especially S. aureus infections, and this article includes design strategies, representative case studies, advantages, limitations, and perspectives to guide future research.
Collapse
Affiliation(s)
- Shuaishuai Ni
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baoli Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Fei Mao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Lefu Lan
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai, China
| | - Jin Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
21
|
Okombo J, Brunschwig C, Singh K, Dziwornu GA, Barnard L, Njoroge M, Wittlin S, Chibale K. Antimalarial Pyrido[1,2- a]benzimidazole Derivatives with Mannich Base Side Chains: Synthesis, Pharmacological Evaluation, and Reactive Metabolite Trapping Studies. ACS Infect Dis 2019; 5:372-384. [PMID: 30608648 DOI: 10.1021/acsinfecdis.8b00279] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A novel series of pyrido[1,2- a]benzimidazoles bearing Mannich base side chains and their metabolites were synthesized and evaluated for in vitro antiplasmodium activity, microsomal metabolic stability, reactive metabolite (RM) formation, and in vivo antimalarial efficacy in a mouse model. Oral administration of one of the derivatives at 4 × 50 mg/kg reduced parasitemia by 95% in Plasmodium berghei-infected mice, with a mean survival period of 16 days post-treatment. The in vivo efficacy of these derivatives is likely a consequence of their active metabolites, two of which showed potent in vitro antiplasmodium activity against chloroquine-sensitive and multidrug-resistant Plasmodium falciparum ( P. falciparum) strains. Rapid metabolism was observed for all the analogues with <40% of parent compound remaining after 30 min of incubation in liver microsomes. RM trapping studies detected glutathione adducts only in derivatives bearing 4-aminophenol moiety, with fragmentation signatures showing that this conjugation occurred on the phenyl ring of the Mannich base side chain. As with amodiaquine (AQ), interchanging the positions of the 4-hydroxyl and Mannich base side group or substituting the 4-hydroxyl with fluorine appeared to block bioactivation of the AQ-like derivatives though at the expense of antiplasmodium activity, which was significantly lowered.
Collapse
Affiliation(s)
- John Okombo
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Christel Brunschwig
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Kawaljit Singh
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Linley Barnard
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, Basel 4002, Switzerland
- University of Basel, Basel 4003, Switzerland
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
22
|
Mayoka G, Keiser J, Häberli C, Chibale K. Structure-Activity Relationship and in Vitro Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) Studies of N-aryl 3-Trifluoromethyl Pyrido[1,2- a]benzimidazoles That Are Efficacious in a Mouse Model of Schistosomiasis. ACS Infect Dis 2019; 5:418-429. [PMID: 30580519 DOI: 10.1021/acsinfecdis.8b00313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We have previously reported on the antischistosomal activity of pyrido[1,2- a]benzimidazole (PBI) derivatives. As a follow-up, we designed and prosecuted further structure-activity relationship (SAR) studies that incorporate N-aryl substitutions on the PBI scaffold. Investigations into the in vitro antischistosomal activity against newly transformed schistosomula (NTS) and adult worms revealed several leads with promising potency. Active compounds with a good cytotoxicity profile were tested in vivo whereby 6 and 44 induced noteworthy reduction (62-69%) in the worm load in the Schistosoma mansoni mouse model. Pharmacokinetic analysis on 44 pointed to slow absorption, low volume of distribution, and low plasma clearance indicating the potential of these compounds to achieve a long duration of action. Overall, our work demonstrates that PBI chemotype is a promising scaffold in the discovery of new antischistosomal leads.
Collapse
Affiliation(s)
- Godfrey Mayoka
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, P.O. Box CH-4003, Basel, Switzerland
| | - Cécile Häberli
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, P.O. Box CH-4003, Basel, Switzerland
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery Unit, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
23
|
Rylands LI, Welsh A, Maepa K, Stringer T, Taylor D, Chibale K, Smith GS. Structure-activity relationship studies of antiplasmodial cyclometallated ruthenium(II), rhodium(III) and iridium(III) complexes of 2-phenylbenzimidazoles. Eur J Med Chem 2019; 161:11-21. [DOI: 10.1016/j.ejmech.2018.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
|
24
|
Gemma S, Federico S, Brogi S, Brindisi M, Butini S, Campiani G. Dealing with schistosomiasis: Current drug discovery strategies. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2019. [DOI: 10.1016/bs.armc.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
25
|
Mayoka G, Njoroge M, Okombo J, Gibhard L, Sanches-Vaz M, Fontinha D, Birkholtz LM, Reader J, van der Watt M, Coetzer TL, Lauterbach S, Churchyard A, Bezuidenhout B, Egan TJ, Yeates C, Wittlin S, Prudêncio M, Chibale K. Structure–Activity Relationship Studies and Plasmodium Life Cycle Profiling Identifies Pan-Active N-Aryl-3-trifluoromethyl Pyrido[1,2-a]benzimidazoles Which Are Efficacious in an in Vivo Mouse Model of Malaria. J Med Chem 2018; 62:1022-1035. [DOI: 10.1021/acs.jmedchem.8b01769] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Godfrey Mayoka
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - John Okombo
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Liezl Gibhard
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Margarida Sanches-Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Theresa L. Coetzer
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Sonja Lauterbach
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Alisje Churchyard
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Belinda Bezuidenhout
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg 2193, South Africa
| | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Clive Yeates
- Inpharma
Consultancy, 6 Dudley Hill Close, Welwyn, Hertfordshire AL60QQ, U.K
| | - Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council, Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
26
|
L'abbate FP, Müller R, Openshaw R, Combrinck JM, de Villiers KA, Hunter R, Egan TJ. Hemozoin inhibiting 2-phenylbenzimidazoles active against malaria parasites. Eur J Med Chem 2018; 159:243-254. [PMID: 30296683 DOI: 10.1016/j.ejmech.2018.09.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/22/2018] [Accepted: 09/24/2018] [Indexed: 12/23/2022]
Abstract
The 2-phenylbenzimidazole scaffold has recently been discovered to inhibit β-hematin (synthetic hemozoin) formation by high throughput screening. Here, a library of 325,728 N-4-(1H-benzo[d]imidazol-2-yl)aryl)benzamides was enumerated, and Bayesian statistics used to predict β-hematin and Plasmodium falciparum growth inhibition. Filtering predicted inactives and compounds with negligible aqueous solubility reduced the library to 35,124. Further narrowing to compounds with terminal aryl ring substituents only, reduced the library to 18, 83% of which were found to inhibit β-hematin formation <100 μM and 50% parasite growth <2 μM. Four compounds showed nanomolar parasite growth inhibition activities, no cross-resistance in a chloroquine resistant strain and low cytotoxicity. QSAR analysis showed a strong association of parasite growth inhibition with inhibition of β-hematin formation and the most active compound inhibited hemozoin formation in P. falciparum, with consequent increasing exchangeable heme. Pioneering use of molecular docking for this system demonstrated predictive ability and could rationalize observed structure activity trends.
Collapse
Affiliation(s)
- Fabrizio P L'abbate
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Ronel Müller
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Roxanne Openshaw
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Jill M Combrinck
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Observatory, 7925, South Africa
| | - Katherine A de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Roger Hunter
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa.
| |
Collapse
|
27
|
Kandepedu N, Gonzàlez Cabrera D, Eedubilli S, Taylor D, Brunschwig C, Gibhard L, Njoroge M, Lawrence N, Paquet T, Eyermann CJ, Spangenberg T, Basarab GS, Street LJ, Chibale K. Identification, Characterization, and Optimization of 2,8-Disubstituted-1,5-naphthyridines as Novel Plasmodium falciparum Phosphatidylinositol-4-kinase Inhibitors with in Vivo Efficacy in a Humanized Mouse Model of Malaria. J Med Chem 2018; 61:5692-5703. [DOI: 10.1021/acs.jmedchem.8b00648] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Nishanth Kandepedu
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Diego Gonzàlez Cabrera
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Srinivas Eedubilli
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Dale Taylor
- Drug Discovery and Development Centre (H3D), DMPK/Pharmacology, University of Cape Town, Observatory 7925, South Africa
| | - Christel Brunschwig
- Drug Discovery and Development Centre (H3D), DMPK/Pharmacology, University of Cape Town, Observatory 7925, South Africa
| | - Liezl Gibhard
- Drug Discovery and Development Centre (H3D), DMPK/Pharmacology, University of Cape Town, Observatory 7925, South Africa
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), DMPK/Pharmacology, University of Cape Town, Observatory 7925, South Africa
| | - Nina Lawrence
- Drug Discovery and Development Centre (H3D), DMPK/Pharmacology, University of Cape Town, Observatory 7925, South Africa
| | - Tanya Paquet
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Charles J. Eyermann
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Thomas Spangenberg
- Merck Global Health Institute, Ares Trading S.A., a subsidiary of Merck KGaA (Darmstadt, Germany), Coinsins 1267, Switzerland
| | - Gregory S. Basarab
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Leslie J. Street
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council, Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
28
|
Antimalarial agents against both sexual and asexual parasites stages: structure-activity relationships and biological studies of the Malaria Box compound 1-[5-(4-bromo-2-chlorophenyl)furan-2-yl]-N-[(piperidin-4-yl)methyl]methanamine (MMV019918) and analogues. Eur J Med Chem 2018; 150:698-718. [PMID: 29571157 DOI: 10.1016/j.ejmech.2018.03.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 01/31/2023]
Abstract
Therapies addressing multiple stages of Plasmodium falciparum life cycle are highly desirable for implementing malaria elimination strategies. MMV019918 (1, 1-[5-(4-bromo-2-chlorophenyl)furan-2-yl]-N-[(piperidin-4-yl)methyl]methanamine) was selected from the MMV Malaria Box for its dual activity against both asexual stages and gametocytes. In-depth structure-activity relationship studies and cytotoxicity evaluation led to the selection of 25 for further biological investigation. The potential transmission blocking activity of 25 versus P. falciparum was confirmed through the standard membrane-feeding assay. Both 1 and 25 significantly prolonged atrioventricular conduction time in Langendorff-isolated rat hearts, and showed inhibitory activity of Ba2+ current through Cav1.2 channels. An in silico target-fishing study suggested the enzyme phosphoethanolamine methyltransferase (PfPMT) as a potential target. However, compound activity against PfPMT did not track with the antiplasmodial activity, suggesting the latter activity relies on a different molecular target. Nevertheless, 25 showed interesting activity against PfPMT, which could be an important starting point for the identification of more potent inhibitors active against both sexual and asexual stages of the parasite.
Collapse
|
29
|
Chen W, Huang Z, Wang W, Mao F, Guan L, Tang Y, Jiang H, Li J, Huang J, Jiang L, Zhu J. Discovery of new antimalarial agents: Second-generation dual inhibitors against FP-2 and PfDHFR via fragments assembely. Bioorg Med Chem 2017; 25:6467-6478. [DOI: 10.1016/j.bmc.2017.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/07/2017] [Accepted: 10/16/2017] [Indexed: 02/02/2023]
|
30
|
Okombo J, Chibale K. Insights into Integrated Lead Generation and Target Identification in Malaria and Tuberculosis Drug Discovery. Acc Chem Res 2017. [PMID: 28636311 PMCID: PMC5518282 DOI: 10.1021/acs.accounts.6b00631] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
New, safe and effective drugs are urgently needed to treat and control malaria and tuberculosis, which affect millions of people annually. However, financial return on investment in the poor settings where these diseases are mostly prevalent is very minimal to support market-driven drug discovery and development. Moreover, the imminent loss of therapeutic lifespan of existing therapies due to evolution and spread of drug resistance further compounds the urgency to identify novel effective drugs. However, the advent of new public-private partnerships focused on tropical diseases and the recent release of large data sets by pharmaceutical companies on antimalarial and antituberculosis compounds derived from phenotypic whole cell high throughput screening have spurred renewed interest and opened new frontiers in malaria and tuberculosis drug discovery. This Account recaps the existing challenges facing antimalarial and antituberculosis drug discovery, including limitations associated with experimental animal models as well as biological complexities intrinsic to the causative pathogens. We enlist various highlights from a body of work within our research group aimed at identifying and characterizing new chemical leads, and navigating these challenges to contribute toward the global drug discovery and development pipeline in malaria and tuberculosis. We describe a catalogue of in-house efforts toward deriving safe and efficacious preclinical drug development candidates via cell-based medicinal chemistry optimization of phenotypic whole-cell medium and high throughput screening hits sourced from various small molecule chemical libraries. We also provide an appraisal of target-based screening, as invoked in our laboratory for mechanistic evaluation of the hits generated, with particular focus on the enzymes within the de novo pyrimidine biosynthetic and hemoglobin degradation pathways, the latter constituting a heme detoxification process and an associated cysteine protease-mediated hydrolysis of hemoglobin. We further expound on the recombinant enzyme assays, heme fractionation experiments, and genomic and chemoproteomic methods that we employed to identify Plasmodium falciparum falcipain 2 (PfFP2), hemozoin formation, phosphatidylinositol 4-kinase (PfPI4K) and Mycobacterium tuberculosis cytochrome bc1 complex as the targets of the antimalarial chalcones, pyrido[1,2-a]benzimidazoles, aminopyridines, and antimycobacterial pyrrolo[3,4-c]pyridine-1,3(2H)-diones, respectively. In conclusion, we argue for the expansion of chemical space through exploitation of privileged natural product scaffolds and diversity-oriented synthesis, as well as the broadening of druggable spaces by exploiting available protein crystal structures, -omics data, and bioinformatics infrastructure to explore hitherto untargeted spaces like lipid metabolism and protein kinases in P. falciparum. Finally, we audit the merits of both target-based and whole-cell phenotypic screening in steering antimalarial and antituberculosis chemical matter toward populating drug discovery pipelines with new lead molecules.
Collapse
Affiliation(s)
- John Okombo
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South
African Medical Research Council Drug Discovery and Development Research
Unit, Drug Discovery and Development Centre (H3D), Department of Chemistry
and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
31
|
Okombo J, Singh K, Mayoka G, Ndubi F, Barnard L, Njogu PM, Njoroge M, Gibhard L, Brunschwig C, Vargas M, Keiser J, Egan TJ, Chibale K. Antischistosomal Activity of Pyrido[1,2-a]benzimidazole Derivatives and Correlation with Inhibition of β-Hematin Formation. ACS Infect Dis 2017; 3:411-420. [PMID: 28440625 DOI: 10.1021/acsinfecdis.6b00205] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The extensive use of praziquantel against schistosomiasis raises concerns about drug resistance. New therapeutic alternatives targeting critical pathways within the parasite are therefore urgently needed. Hemozoin formation in Schistosoma presents one such target. We assessed the in vitro antischistosomal activity of pyrido[1,2-a]benzimidazoles (PBIs) and investigated correlations with their ability to inhibit β-hematin formation. We further evaluated the in vivo efficacy of representative compounds in experimental mice and conducted pharmacokinetic analysis on the most potent. At 10 μM, 48/57 compounds resulted in >70% mortality of newly transformed schistosomula, whereas 37 of these maintained >60% mortality of adult S. mansoni. No correlations were observed between β-hematin inhibitory and antischistosomal activities against both larval and adult parasites, suggesting possible presence of other target(s) or a mode of inhibition of crystal formation that is not adequately modeled by the assay. The most active compound in vivo showed 58.7 and 61.3% total and female worm burden reduction, respectively. Pharmacokinetic analysis suggested solubility-limited absorption and high hepatic clearance as possible contributors to the modest efficacy despite good in vitro activity. The PBIs evaluated in this report thus merit further optimization to improve their efficacy and to elucidate their possible mode of action.
Collapse
Affiliation(s)
- John Okombo
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kawaljit Singh
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Godfrey Mayoka
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Ferdinand Ndubi
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Linley Barnard
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Peter M. Njogu
- Department of Pharmaceutical Chemistry, University of Nairobi, P.O. Box 19676, Nairobi 00202, Kenya
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D),
Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Liezl Gibhard
- Drug Discovery and Development Centre (H3D),
Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Christel Brunschwig
- Drug Discovery and Development Centre (H3D),
Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Mireille Vargas
- Department of Medical Parasitology and
Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
- University of Basel, 4003 Basel, Switzerland
| | - Jennifer Keiser
- Department of Medical Parasitology and
Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
- University of Basel, 4003 Basel, Switzerland
| | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council,
Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|