1
|
Ishmail FZ, Coertzen D, Tshabalala S, Leshabane M, da Rocha S, Njoroge M, Gibhard L, Birkholtz LM, Woodland JG, Egan TJ, Wicht KJ, Chibale K. Synthesis and SAR Studies of Acyl-Thiourea Platinum(II) Complexes Yield Analogs with Dual-Stage Antiplasmodium Activity. ACS Med Chem Lett 2025; 16:428-435. [PMID: 40104806 PMCID: PMC11912270 DOI: 10.1021/acsmedchemlett.4c00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Mixed-ligand platinum(II) complexes incorporating bipyridine and acyl-thiourea ligands were synthesized and evaluated for their in vitro growth inhibitory activity against the human malaria parasite Plasmodium falciparum (Pf). The substituents at four distinct sites were varied to identify structure-activity relationships for this series. Most complexes displayed potent PfNF54 activity with IC50 values in the nanomolar range and favorable cytotoxicity profiles. Five complexes (C1, C11, C12, C15, and C17) exhibited activity against both the asexual blood and sexual (gametocyte) stage parasites, with another complex (C8) exhibiting activity against late-stage gametocytes only. In addition, the complexes showed comparable ABS potency against the PfK1 multidrug-resistant strain. The pharmacokinetic parameters of one analog (C6), which displayed good solubility and mouse microsomal metabolic stability, were measured. This work demonstrates the potential of acyl-thiourea platinum(II) complexes as selective, multistage-active antiplasmodium compounds as part of the search for new antimalarial agents.
Collapse
Affiliation(s)
| | - Dina Coertzen
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Hatfield 0028, South Africa
| | - Sizwe Tshabalala
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Hatfield 0028, South Africa
| | - Meta Leshabane
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Hatfield 0028, South Africa
| | - Shante da Rocha
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Hatfield 0028, South Africa
| | - Mathew Njoroge
- Holistic
Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch 7701, South Africa
| | - Liezl Gibhard
- Holistic
Drug Discovery and Development (H3D) Centre, 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, Hatfield 0028, South Africa
- Institute
for Sustainable Malaria Control, School of Public Health and Health
Systems, University of Pretoria, Hatfield 0028, South Africa
- Department
of Biochemistry, Stellenbosch University, Stellenbosch, Matieland 7602, South Africa
| | - John G. Woodland
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Holistic
Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch 7701, South Africa
- South
African
Medical Research Council Drug Discovery and Development Research Unit,
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Timothy J. Egan
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South
African
Medical Research Council Drug Discovery and Development Research Unit,
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Kathryn J. Wicht
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Holistic
Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch 7701, South Africa
- South
African
Medical Research Council Drug Discovery and Development Research Unit,
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Kelly Chibale
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Holistic
Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch 7701, South Africa
- South
African
Medical Research Council Drug Discovery and Development Research Unit,
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa
| |
Collapse
|
2
|
Ul Haq I, Pinto Vieira R, Lima WG, de Lima ME, Krukiewicz K. Antimicrobial polymers: elucidating the role of functional groups on antimicrobial activity. ARAB JOURNAL OF BASIC AND APPLIED SCIENCES 2024; 31:325-344. [DOI: 10.1080/25765299.2024.2366543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland
- Joint Doctoral School, Silesian University of Technology, Gliwice, Poland
- Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rafael Pinto Vieira
- Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - William Gustavo Lima
- Programa de Pós-Graduação Stricto Sensu em Medicina e Biomedicina da Faculdade, Santa Casa de Belo Horizonte, Belo Horizonte, MG, Brazil
| | - Maria Elena de Lima
- Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Programa de Pós Graduação em Medicina e Biomedicina da Faculdade de Saúde, Santa Casa de Belo Horizonte, Belo Horizonte, MG, Brazil
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
3
|
Watson SJ, van der Watt ME, Theron A, Reader J, Tshabalala S, Erlank E, Koekemoer LL, Naude M, Stampolaki M, Adewole F, Sadowska K, Pérez-Lozano P, Turcu AL, Vázquez S, Ko J, Mazurek B, Singh D, Malwal SR, Njoroge M, Chibale K, Onajole OK, Kolocouris A, Oldfield E, Birkholtz LM. The Tuberculosis Drug Candidate SQ109 and Its Analogs Have Multistage Activity against Plasmodium falciparum. ACS Infect Dis 2024; 10:3358-3367. [PMID: 39143042 PMCID: PMC11406516 DOI: 10.1021/acsinfecdis.4c00461] [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] [Indexed: 08/16/2024]
Abstract
Toward repositioning the antitubercular clinical candidate SQ109 as an antimalarial, analogs were investigated for structure-activity relationships for activity against asexual blood stages of the human malaria parasite Plasmodium falciparum pathogenic forms, as well as transmissible, sexual stage gametocytes. We show that equipotent activity (IC50) in the 100-300 nM range could be attained for both asexual and sexual stages, with the activity of most compounds retained against a multidrug-resistant strain. The multistage activity profile relies on high lipophilicity ascribed to the adamantane headgroup, and antiplasmodial activity is critically dependent on the diamine linker. Frontrunner compounds showed conserved activity against genetically diverse southern African clinical isolates. We additionally validated that this series could block transmission to mosquitoes, marking these compounds as novel chemotypes with multistage antiplasmodial activity.
Collapse
Affiliation(s)
| | | | - Anjo Theron
- Next Generation Health, Council for Scientific and Industrial Research, Pretoria 0001, South Africa
| | | | | | - Erica Erlank
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Services, Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Johannesburg 2000, South Africa
| | - Lizette L Koekemoer
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Services, Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Johannesburg 2000, South Africa
| | | | - Marianna Stampolaki
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis-Zografou, 15771 Athens, Greece
| | - Feyisola Adewole
- Department of Biological, Physical and Health Sciences, College of Science, Health & Pharmacy, Roosevelt University, 425 South Wabash Avenue, Chicago, Illinois 60605, United States
| | - Katie Sadowska
- Department of Biological, Physical and Health Sciences, College of Science, Health & Pharmacy, Roosevelt University, 425 South Wabash Avenue, Chicago, Illinois 60605, United States
| | - Pilar Pérez-Lozano
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain
| | - Andreea L Turcu
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona E-08028, Spain
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona E-08028, Spain
| | - Jihee Ko
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Ben Mazurek
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Davinder Singh
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Satish R Malwal
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Capetown 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Capetown 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Centre, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Capetown 7701, South Africa
| | - Oluseye K Onajole
- Department of Biological, Physical and Health Sciences, College of Science, Health & Pharmacy, Roosevelt University, 425 South Wabash Avenue, Chicago, Illinois 60605, United States
| | - Antonios Kolocouris
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis-Zografou, 15771 Athens, Greece
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | | |
Collapse
|
4
|
Bagratee T, Prawlall R, Ndlovu T, Sibisi S, Ndadane S, Shaik BB, Palkar MB, Gampa R, Karpoormath R. Exploring the Recent Pioneering Developments of Small Molecules in Antimalarial Drug Armamentarium: A Chemistry Prospective Appraisal. Chem Biodivers 2024; 21:e202400460. [PMID: 38759144 DOI: 10.1002/cbdv.202400460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/19/2024]
Abstract
Malaria is a very destructive and lethal parasitic disease that causes significant mortality worldwide, resulting in the loss of millions of lives annually. It is an infectious disease transmitted by mosquitoes, which is caused by different species of the parasite protozoan belonging to the genus Plasmodium. The uncontrolled intake of antimalarial drugs often employed in clinical settings has resulted in the emergence of numerous strains of plasmodium that are resistant to these drugs, including multidrug-resistant strains. This resistance significantly diminishes the effectiveness of many primary drugs used in the treatment of malaria. Hence, there is an urgent need for developing unique classes of antimalarial drugs that function with distinct mechanisms of action. In this context, the design and development of hybrid compounds that combine pharmacophoric properties from different lead molecules into a single unit gives a unique perspective towards further development of malaria drugs in the next generation. In recent years, the field of medicinal chemistry has made significant efforts resulting in the discovery and synthesis of numerous small novel compounds that exhibit potent antimalarial properties, while also demonstrating reduced toxicity and desirable efficacy. In light of this, we have reviewed the progress of hybrid antimalarial agents from 2021 up to the present. This manuscript presents a comprehensive overview of the latest advancements in the medicinal chemistry pertaining to small molecules, with a specific focus on their potential as antimalarial agents. As possible antimalarial drugs that might target both the dual stage and multi-stage stages of the parasite life cycle, these small hybrid molecules have been studied. This review explores a variety of physiologically active compounds that have been described in the literature in order to lay a strong foundation for the logical design and eventual identification of antimalarial drugs based on lead frameworks.
Collapse
Affiliation(s)
- Tameika Bagratee
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Ritika Prawlall
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Thabani Ndlovu
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Sinqobile Sibisi
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Sisa Ndadane
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Baji Baba Shaik
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Mahesh B Palkar
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
- Department of Pharmaceutical Chemistry, SVKM's NMIMS, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Raghavachary Gampa
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal (Westville), Durban, 4000, South Africa
| |
Collapse
|
5
|
Krstulović L, Rastija V, Pessanha de Carvalho L, Held J, Rajić Z, Živković Z, Bajić M, Glavaš-Obrovac L. Design, Synthesis, Antitumor, and Antiplasmodial Evaluation of New 7-Chloroquinoline-Benzimidazole Hybrids. Molecules 2024; 29:2997. [PMID: 38998949 PMCID: PMC11243327 DOI: 10.3390/molecules29132997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Newly synthesized 7-chloro-4-aminoquinoline-benzimidazole hybrids were characterized by NMR and elemental analysis. Compounds were tested for their effects on the growth of the non-tumor cell line MRC-5 (human fetal lung fibroblasts) and carcinoma (HeLa and CaCo-2), leukemia, and lymphoma (Hut78, THP-1, and HL-60) cell lines. The obtained results, expressed as the concentration at which 50% inhibition of cell growth is achieved (IC50 value), show that the tested compounds affect cell growth differently depending on the cell line and the applied dose (IC50 ranged from 0.2 to >100 µM). Also, the antiplasmodial activity of these hybrids was evaluated against two P. falciparum strains (Pf3D7 and PfDd2). The tested compounds showed potent antiplasmodial activity, against both strains, at nanomolar concentrations. Quantitative structure-activity relationship (QSAR) analysis resulted in predictive models for antiplasmodial activity against the 3D7 strain (R2 = 0.886; Rext2 = 0.937; F = 41.589) and Dd2 strain (R2 = 0.859; Rext2 = 0.878; F = 32.525) of P. falciparum. QSAR models identified the structural features of these favorable effects on antiplasmodial activities.
Collapse
Affiliation(s)
- Luka Krstulović
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, HR-10000 Zagreb, Croatia;
| | - Vesna Rastija
- Department of Agroecology and Environmental Protection, Faculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek, Vladimira Preloga 1, HR-31000 Osijek, Croatia;
| | - Lais Pessanha de Carvalho
- Institute of Tropical Medicine, University of Tuebingen, Wilhelmstrasse 27, D-72074 Tuebingen, Germany; (L.P.d.C.); (J.H.)
| | - Jana Held
- Institute of Tropical Medicine, University of Tuebingen, Wilhelmstrasse 27, D-72074 Tuebingen, Germany; (L.P.d.C.); (J.H.)
- Partner Site Tuebingen, German Center for Infection Research (DZIF),Wilhelmstrasse 27, D-72074 Tuebingen, Germany
| | - Zrinka Rajić
- Department of Medicinal Chemistry, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, HR-10000 Zagreb, Croatia;
| | - Zorislava Živković
- General County Hospital of Našice, Bana Jelačića 10, HR-31500 Našice, Croatia;
| | - Miroslav Bajić
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, HR-10000 Zagreb, Croatia;
| | - Ljubica Glavaš-Obrovac
- Department of Medicinal Chemistry, Biochemistry, and Clinical Chemistry, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| |
Collapse
|
6
|
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
|
7
|
Brenda CT, Norma RF, P BN, E CR, Nelly LV, Marcela RL, Martha UC, I FT. Ultrastructural alterations due to sodium metavanadate treatment in the blood stages of Plasmodium yoelii yoelii. J Trace Elem Med Biol 2023; 80:127314. [PMID: 37778096 DOI: 10.1016/j.jtemb.2023.127314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023]
Abstract
Malaria is a potentially mortal disease caused by parasites of the genus Plasmodium spp. It has a wide distribution in the world and unfortunately there are several factors that make its control difficult; among which the development of pharmacological resistance to the different drugs used to treat this disease stands out, which makes it necessary to design new compounds that have an antimalarial effect. Previous studies have shown that vanadium has a broad antiparasitic spectrum and is also safe for the host, so the objective of this research was to evaluate the antimalarial potential of sodium metavanadate (SM) and to analyze the ultrastructural changes in parasites exposed. The method consisted of inoculating CD-1 male mice with Plasmodium yoelii yoelii and administering a 10 mg/kg/day dose of SM orally for 4 days. On the fifth day, whole blood samples were obtained, processed for ultrastructural analysis, and the changes in the different parasite stages were compared against the control. Our results showed that SM decreased parasitemia compared to the group that did not receive treatment and modified the ultrastructure in all parasitic stages because it damaged the membranes, causing alterations mainly in the nucleus and in the mitochondria as well as the loss of cellular organization, which could affect the integrity of these parasites and decrease its viability.
Collapse
Affiliation(s)
- Casarrubias-Tabarez Brenda
- Department of Cellular and Tissular Biology, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C.P. 04510, Mexico
| | - Rivera-Fernández Norma
- Department of Microbiology and Parasitology. School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico
| | - Bizarro-Nevares P
- Department of Cellular and Tissular Biology, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico
| | - Carrasco-Ramírez E
- Department of Microbiology and Parasitology. School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico; Microscopy Unit, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico
| | - López-Valdez Nelly
- Department of Cellular and Tissular Biology, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico
| | - Rojas-Lemus Marcela
- Department of Cellular and Tissular Biology, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico
| | - Ustarroz-Cano Martha
- Department of Cellular and Tissular Biology, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico
| | - Fortoul Teresa I
- Department of Cellular and Tissular Biology, School of Medicine, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, Mexico City C.P. 04510, Mexico.
| |
Collapse
|
8
|
Hikaambo CN, Shakela N, Woodland JG, Wicht KJ, Chibale K. Drug discovery in Africa tackles zoonotic and related infections. Sci Transl Med 2023; 15:eadj0035. [PMID: 37851825 DOI: 10.1126/scitranslmed.adj0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Zoonotic and related infections pose an enormous health threat to the world's second-most populous continent. Despite the challenges faced by drug discovery scientists in Africa, recent progress toward identifying potential medicines across diverse disease areas is a cause for optimism and an indicator of progress in African-led scientific initiatives.
Collapse
Affiliation(s)
- Christabel N Hikaambo
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Natalia Shakela
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - John G Woodland
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| | - Kathryn J Wicht
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, Cape Town, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| |
Collapse
|
9
|
Shen S, Xu W, Lu J, Wang S, Huang Y, Zeng X, Xiao W, Yin J. Recent progress on fluorescent probes for viruses. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
10
|
Stampolaki M, Malwal SR, Alvarez-Cabrera N, Gao Z, Moniruzzaman M, Babii SO, Naziris N, Rey-Cibati A, Valladares-Delgado M, Turcu AL, Baek KH, Phan TN, Lee H, Alcaraz M, Watson S, van der Watt M, Coertzen D, Efstathiou N, Chountoulesi M, Shoen CM, Papanastasiou IP, Brea J, Cynamon MH, Birkholtz LM, Kremer L, No JH, Vázquez S, Benaim G, Demetzos C, Zgurskaya HI, Dick T, Oldfield E, D. Kolocouris A. Synthesis and Testing of Analogs of the Tuberculosis Drug Candidate SQ109 against Bacteria and Protozoa: Identification of Lead Compounds against Mycobacterium abscessus and Malaria Parasites. ACS Infect Dis 2023; 9:342-364. [PMID: 36706233 PMCID: PMC10615177 DOI: 10.1021/acsinfecdis.2c00537] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
SQ109 is a tuberculosis drug candidate that has high potency against Mycobacterium tuberculosis and is thought to function at least in part by blocking cell wall biosynthesis by inhibiting the MmpL3 transporter. It also has activity against bacteria and protozoan parasites that lack MmpL3, where it can act as an uncoupler, targeting lipid membranes and Ca2+ homeostasis. Here, we synthesized 18 analogs of SQ109 and tested them against M. smegmatis, M. tuberculosis, M. abscessus, Bacillus subtilis, and Escherichia coli, as well as against the protozoan parasites Trypanosoma brucei, T. cruzi, Leishmania donovani, L. mexicana, and Plasmodium falciparum. Activity against the mycobacteria was generally less than with SQ109 and was reduced by increasing the size of the alkyl adduct, but two analogs were ∼4-8-fold more active than SQ109 against M. abscessus, including a highly drug-resistant strain harboring an A309P mutation in MmpL3. There was also better activity than found with SQ109 with other bacteria and protozoa. Of particular interest, we found that the adamantyl C-2 ethyl, butyl, phenyl, and benzyl analogs had 4-10× increased activity against P. falciparum asexual blood stages, together with low toxicity to a human HepG2 cell line, making them of interest as new antimalarial drug leads. We also used surface plasmon resonance to investigate the binding of inhibitors to MmpL3 and differential scanning calorimetry to investigate binding to lipid membranes. There was no correlation between MmpL3 binding and M. tuberculosis or M. smegmatis cell activity, suggesting that MmpL3 is not a major target in mycobacteria. However, some of the more active species decreased lipid phase transition temperatures, indicating increased accumulation in membranes, which is expected to lead to enhanced uncoupler activity.
Collapse
Affiliation(s)
- Marianna Stampolaki
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - Satish R. Malwal
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, USA
| | | | - Zijun Gao
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, USA
| | - Mohammad Moniruzzaman
- University of Oklahoma, Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, OK 73019-5251, USA
| | - Svitlana O. Babii
- University of Oklahoma, Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, OK 73019-5251, USA
| | - Nikolaos Naziris
- Section of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - André Rey-Cibati
- Instituto de Estudios Avanzados, Caracas, Venezuela Instituto de Biología Experimental, Facultad de Ciencias, Universidad Central de Venezuela (UCV), Caracas, Venezuela
| | - Mariana Valladares-Delgado
- Instituto de Estudios Avanzados, Caracas, Venezuela Instituto de Biología Experimental, Facultad de Ciencias, Universidad Central de Venezuela (UCV), Caracas, Venezuela
| | - Andreea L. Turcu
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, Barcelona, E-08028, Spain
| | - Kyung-Hwa Baek
- Host-Parasite Research Laboratory, Institut Pasteur Korea, Seongnam-si, Republic of Korea
| | - Trong-Nhat Phan
- Host-Parasite Research Laboratory, Institut Pasteur Korea, Seongnam-si, Republic of Korea
| | - Hyeryon Lee
- Host-Parasite Research Laboratory, Institut Pasteur Korea, Seongnam-si, Republic of Korea
| | - Mattheo Alcaraz
- Institut de Recherche en Infectiologie de Montpellier, CNRS UMR9004, Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
| | - Savannah Watson
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Mariette van der Watt
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Dina Coertzen
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Natasa Efstathiou
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - Maria Chountoulesi
- Section of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - Carolyn M. Shoen
- Central New York Research Corporation, Veterans Affairs Medical Center, Syracuse, NY 13210, U
| | - Ioannis P. Papanastasiou
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - Jose Brea
- Drug Screening Platform/Biofarma Research Group, CIMUS Research Center, Departamento de Farmacoloxía, Farmacia e Tecnoloxía Farmacéutica, University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain
| | - Michael H. Cynamon
- Central New York Research Corporation, Veterans Affairs Medical Center, Syracuse, NY 13210, U
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Laurent Kremer
- Institut de Recherche en Infectiologie de Montpellier, CNRS UMR9004, Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
- INSERM, IRIM, Montpellier, France
| | - Joo Hwan No
- Host-Parasite Research Laboratory, Institut Pasteur Korea, Seongnam-si, Republic of Korea
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, Barcelona, E-08028, Spain
| | - Gustavo Benaim
- Instituto de Estudios Avanzados, Caracas, Venezuela Instituto de Biología Experimental, Facultad de Ciencias, Universidad Central de Venezuela (UCV), Caracas, Venezuela
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - Helen I. Zgurskaya
- University of Oklahoma, Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, 101 Stephenson Parkway, Norman, OK 73019-5251, USA
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC 20007, USA
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, USA
| | - Antonios D. Kolocouris
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| |
Collapse
|
11
|
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
|
12
|
Reader J, Opperman DFL, van der Watt ME, Theron A, Leshabane M, da Rocha S, Turner J, Garrabrant K, Piña I, Mills C, Woster PM, Birkholtz L. New Transmission-Selective Antimalarial Agents through Hit-to-Lead Optimization of 2-([1,1'-Biphenyl]-4-carboxamido)benzoic Acid Derivatives. Chembiochem 2022; 23:e202200427. [PMID: 36106425 PMCID: PMC10946866 DOI: 10.1002/cbic.202200427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/14/2022] [Indexed: 11/12/2022]
Abstract
Malaria elimination requires multipronged approaches, including the application of antimalarial drugs able to block human-to-mosquito transmission of malaria parasites. The transmissible gametocytes of Plasmodium falciparum seem to be highly sensitive towards epidrugs, particularly those targeting demethylation of histone post-translational marks. Here, we report exploration of compounds from a chemical library generated during hit-to-lead optimization of inhibitors of the human histone lysine demethylase, KDM4B. Derivatives of 2-([1,1'-biphenyl]-4-carboxamido) benzoic acid, around either the amide or a sulfonamide linker backbone (2-(arylcarboxamido)benzoic acid, 2-carboxamide (arylsulfonamido)benzoic acid and N-(2-(1H-tetrazol-5-yl)phenyl)-arylcarboxamide), showed potent activity towards late-stage gametocytes (stage IV/V) of P. falciparum, with the most potent compound reaching single digit nanomolar activity. Structure-activity relationship trends were evident and frontrunner compounds also displayed microsomal stability and favourable solubility profiles. Simplified synthetic routes support further derivatization of these compounds for further development of these series as malaria transmission-blocking agents.
Collapse
Affiliation(s)
- Janette Reader
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Daniel F. L. Opperman
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Mariëtte E. van der Watt
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
- School of Health Systems and Public HealthUniversity of Pretoria, HatfieldPretoria0028South Africa
| | - Anjo Theron
- Next Generation HealthCouncil for Scientific and Industrial ResearchPretoria0001South Africa
| | - Meta Leshabane
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Shanté da Rocha
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| | - Jonathan Turner
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Kathleen Garrabrant
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Ivett Piña
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Catherine Mills
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Patrick M. Woster
- Department of Drug Discovery and Biomedical SciencesMedical University of South CarolinaCharlestonSC 29425USA
| | - Lyn‐Marié Birkholtz
- Department of BiochemistryGenetics and MicrobiologyInstitute for Sustainable Malaria ControlUniversity of PretoriaLynnwood RoadPretoria0028South Africa
| |
Collapse
|
13
|
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
|
14
|
Chedupaka R, Pawar R, Venkatesham P, Vedula RR. Synthesis, characterization and Density Functional Theory of novel one-pot thioalkylated benzimidazole-linked 4-substituted mercaptoimidazole molecular hybrids via multi-component approach. SYNTHETIC COMMUN 2022. [DOI: 10.1080/00397911.2022.2072745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Raju Chedupaka
- Department of chemistry, National Institute of Technology Warangal, Warangal, India
| | - Ravinder Pawar
- Department of chemistry, National Institute of Technology Warangal, Warangal, India
| | | | - Rajeswar Rao Vedula
- Department of chemistry, National Institute of Technology Warangal, Warangal, India
| |
Collapse
|
15
|
Synthesis and study of organometallic PGM complexes containing 2-(2-pyridyl)benzimidazole as antiplasmodial agents. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
16
|
Consalvi S, Tammaro C, Appetecchia F, Biava M, Poce G. Malaria transmission blocking compounds: a patent review. Expert Opin Ther Pat 2022; 32:649-666. [PMID: 35240899 DOI: 10.1080/13543776.2022.2049239] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite substantial progress in the field, malaria remains a global health issue and currently available control strategies are not sufficient to achieve eradication. Agents able to prevent transmission are likely to have a strong impact on malaria control and have been prioritized as a primary objective to reduce the number of secondary infections. Therefore, there is an increased interest in finding novel drugs targeting sexual stages of Plasmodium and innovative methods to target malaria transmission from host to vector, and vice versa. AREAS COVERED This review covers innovative transmission-blocking inventions patented between 2015 and October 2021. The focus is on chemical interventions which could be used as "chemical vaccines" to prevent transmission (small molecules, carbohydrates, and polypeptides). EXPERT OPINION Even though the development of novel strategies to block transmission still requires fundamental additional research and a deeper understanding of parasite sexual stages biology, the research in this field has significantly accelerated. Among innovative inventions patented over the last six years, the surface-delivery of antimalarial drugs to kill transmission-stages parasites in mosquitoes holds the highest promise for success in malaria control strategies, opening completely new scenarios in malaria transmission-blocking drug discovery.
Collapse
Affiliation(s)
- Sara Consalvi
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Chiara Tammaro
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Federico Appetecchia
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Mariangela Biava
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
17
|
Yu S, Wang J, Luo X, Zheng H, Wang L, Yang X, Wang Y. Transmission-Blocking Strategies Against Malaria Parasites During Their Mosquito Stages. Front Cell Infect Microbiol 2022; 12:820650. [PMID: 35252033 PMCID: PMC8889032 DOI: 10.3389/fcimb.2022.820650] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Malaria is still the most widespread parasitic disease and causes the most infections globally. Owing to improvements in sanitary conditions and various intervention measures, including the use of antimalarial drugs, the malaria epidemic in many regions of the world has improved significantly in the past 10 years. However, people living in certain underdeveloped areas are still under threat. Even in some well-controlled areas, the decline in malaria infection rates has stagnated or the rates have rebounded because of the emergence and spread of drug-resistant malaria parasites. Thus, new malaria control methods must be developed. As the spread of the Plasmodium parasite is dependent on the part of its life cycle that occurs in mosquitoes, to eliminate the possibility of malaria infections, transmission-blocking strategies against the mosquito stage should be the first choice. In fact, after the gametocyte enters the mosquito body, it undergoes a series of transformation processes over a short period, thus providing numerous potential blocking targets. Many research groups have carried out studies based on targeting the blocking of transmission during the mosquito phase and have achieved excellent results. Meanwhile, the direct killing of mosquitoes could also significantly reduce the probability of malaria infections. Microorganisms that display complex interactions with Plasmodium, such as Wolbachia and gut flora, have shown observable transmission-blocking potential. These could be used as a biological control strategy and play an important part in blocking the transmission of malaria.
Collapse
Affiliation(s)
- Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Luo
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Luhan Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
- *Correspondence: Ying Wang,
| |
Collapse
|
18
|
Rational drug design, synthesis, and biological evaluation of novel N-(2-arylaminophenyl)-2,3-diphenylquinoxaline-6-sulfonamides as potential antimalarial, antifungal, and antibacterial agents. DIGITAL CHINESE MEDICINE 2021. [DOI: 10.1016/j.dcmed.2021.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
19
|
Niemand J, van Biljon R, van der Watt M, van Heerden A, Reader J, van Wyk R, Orchard L, Chibale K, Llinás M, Birkholtz LM. Chemogenomic Fingerprints Associated with Stage-Specific Gametocytocidal Compound Action against Human Malaria Parasites. ACS Infect Dis 2021; 7:2904-2916. [PMID: 34569223 DOI: 10.1021/acsinfecdis.1c00373] [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] [Indexed: 11/30/2022]
Abstract
Kinase-focused inhibitors previously revealed compounds with differential activity against different stages of Plasmodium falciparum gametocytes. MMV666810, a 2-aminopyrazine, is more active on late-stage gametocytes, while a pyrazolopyridine, MMV674850, preferentially targets early-stage gametocytes. Here, we probe the biological mechanisms underpinning this differential stage-specific killing using in-depth transcriptome fingerprinting. Compound-specific chemogenomic profiles were observed with MMV674850 treatment associated with biological processes shared between asexual blood stage parasites and early-stage gametocytes but not late-stage gametocytes. MMV666810 has a distinct profile with clustered gene sets associated primarily with late-stage gametocyte development, including Ca2+-dependent protein kinases (CDPK1 and 5) and serine/threonine protein kinases (FIKK). Chemogenomic profiling therefore highlights essential processes in late-stage gametocytes, on a transcriptional level. This information is important to prioritize compounds that preferentially compromise late-stage gametocytes for further development as transmission-blocking antimalarials.
Collapse
Affiliation(s)
- Jandeli Niemand
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Riëtte van Biljon
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Ashleigh van Heerden
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Roelof van Wyk
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Lindsey Orchard
- Department of Biochemistry & Molecular Biology and the Huck Centre for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town, 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
| | - Manuel Llinás
- Department of Biochemistry & Molecular Biology and the Huck Centre for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
20
|
Identification of 3,4-Dihydro-2 H,6 H-pyrimido[1,2- c][1,3]benzothiazin-6-imine Derivatives as Novel Selective Inhibitors of Plasmodium falciparum Dihydroorotate Dehydrogenase. Int J Mol Sci 2021; 22:ijms22137236. [PMID: 34281290 PMCID: PMC8268581 DOI: 10.3390/ijms22137236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Plasmodium falciparum's resistance to available antimalarial drugs highlights the need for the development of novel drugs. Pyrimidine de novo biosynthesis is a validated drug target for the prevention and treatment of malaria infection. P. falciparum dihydroorotate dehydrogenase (PfDHODH) catalyzes the oxidation of dihydroorotate to orotate and utilize ubiquinone as an electron acceptor in the fourth step of pyrimidine de novo biosynthesis. PfDHODH is targeted by the inhibitor DSM265, which binds to a hydrophobic pocket located at the N-terminus where ubiquinone binds, which is known to be structurally divergent from the mammalian orthologue. In this study, we screened 40,400 compounds from the Kyoto University chemical library against recombinant PfDHODH. These studies led to the identification of 3,4-dihydro-2H,6H-pyrimido[1,2-c][1,3]benzothiazin-6-imine and its derivatives as a new class of PfDHODH inhibitor. Moreover, the hit compounds identified in this study are selective for PfDHODH without inhibition of the human enzymes. Finally, this new scaffold of PfDHODH inhibitors showed growth inhibition activity against P. falciparum 3D7 with low toxicity to three human cell lines, providing a new starting point for antimalarial drug development.
Collapse
|