1
|
Nicco E, Lejon V, Mwamba Miaka E, Mumba D, Mpanya A, Kambo C, Ngolo D, Mutombo W, Hugonnet S, Rembry S, Tipple C, Inocencio Da Luz R, Snijders R, Vander Kelen C, Rogé S, Van Reet N, Tarral A, Verlé P, Hasker E. The STROGHAT study protocol: An intervention study to evaluate safety, effectiveness and feasibility of treating gambiense HAT seropositive subjects with acoziborole. OPEN RESEARCH EUROPE 2025; 5:23. [PMID: 40191624 PMCID: PMC11971624 DOI: 10.12688/openreseurope.19077.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 04/09/2025]
Abstract
Background Coordinated efforts in the control of gambiense human African trypanosomiasis (gHAT) have significantly reduced its endemicity. WHO targets interruption of transmission by 2030. However, challenges remain, including low sensitivity of the current parasitological confirmation tests, leaving a potential human reservoir untreated. Acoziborole, a single-dose oral treatment, effective in both disease stages with a good safety profile, offers the potential of treatment of parasitologically negative gHAT seropositive subjects, which could improve diagnostic sensitivity. The STROGHAT study aims to evaluate whether this approach can lead to elimination of T.b. gambiense from its human reservoir, and to provide further safety data on acoziborole. It also includes a costing analysis and a prospective evaluation of the performance of the screening and diagnostic tests used. Methods STROGHAT is a one-arm epidemiological study, with a nested phase IIIb, one-arm, open label, non-randomized, multicentre clinical trial. It will be implemented over four years in the endemic region of the Equateur North, in the Democratic Republic of Congo. For the first three years, parasitologically negative gHAT seropositive subjects will be treated with acoziborole, while parasitologically confirmed cases will receive standard of care. Individual follow-up needs and accurate prevalence estimate will be based on immunological and molecular tests performed for all gHAT screening test seropositive subjects at a reference laboratory. In the fourth year, standard procedures will resume, and a prevalence survey will assess whether interruption of transmission has been achieved. Discussion The manuscript outlines the study background, objectives and methods while discussing its strengths and challenges. If successful, the STROGHAT study will provide critical evidence on the effectiveness, safety and feasibility of the new strategy, and inform future elimination strategies. Clinical trial registration NCT06356974. Date of registration: April 4, 2024.
Collapse
Affiliation(s)
- Elena Nicco
- Institute of Tropical Medicine Antwerp, Antwerp, 2000, Belgium
| | - Veerle Lejon
- French National Research Institute for Sustainable Development IRD, CIRAD, University of Montpellier, Montpellier, 34398, France
| | - Erick Mwamba Miaka
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | - Dieudonné Mumba
- Av. De la Démocratie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Alain Mpanya
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | - Charles Kambo
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | - Digas Ngolo
- Drugs for Neglected Diseases initiative Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Wilfried Mutombo
- Drugs for Neglected Diseases initiative Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Stéphane Hugonnet
- Drugs for Neglected Diseases initiative, Geneva, Geneva, Switzerland
| | - Sandra Rembry
- Drugs for Neglected Diseases initiative, Geneva, Geneva, Switzerland
| | - Craig Tipple
- Drugs for Neglected Diseases initiative, Geneva, Geneva, Switzerland
| | | | - Rian Snijders
- Institute of Tropical Medicine Antwerp, Antwerp, 2000, Belgium
| | | | - Stijn Rogé
- Institute of Tropical Medicine Antwerp, Antwerp, 2000, Belgium
| | - Nick Van Reet
- Institute of Tropical Medicine Antwerp, Antwerp, 2000, Belgium
| | | | - Paul Verlé
- Independent Consultant, Antwerp, 2000, Belgium
| | - Epco Hasker
- Institute of Tropical Medicine Antwerp, Antwerp, 2000, Belgium
| |
Collapse
|
2
|
Ilbeigi K, Mabille D, Matheeussen A, Hendrickx R, Claes M, Van Reet N, Anthonissen R, Hulpia F, Lin C, Maes L, Regnault C, Whitfield P, Roy R, Ungogo MA, Sterckx YGJ, De Winter H, Mertens B, Bundschuh M, De Koning HP, Van Calenbergh S, Caljon G. Discovery and Development of an Advanced Lead for the Treatment of African Trypanosomiasis. ACS Infect Dis 2025; 11:131-143. [PMID: 39665421 DOI: 10.1021/acsinfecdis.4c00472] [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: 12/13/2024]
Abstract
African trypanosomiasis is a widespread disease of human and veterinary importance caused by various Trypanosoma spp. with a globally devastating impact and a need for novel treatment options. We here provide a comprehensive preclinical evaluation of nucleoside analogues, 6-thioether-modified tubercidins, with curative activity against African trypanosomiasis. Promising hits were identified following in vitro screening against the most relevant trypanosome species. Selected hit compounds were extensively tested for in vitro metabolic stability, potency in in vivo mouse models for the various species, genotoxicity in an in vitro testing battery, and mode of action studies (i.e., genome-wide RNA interference library screening and metabolomics). Among the nucleoside analogues, analogue 3 was curative in mouse models with no indication of genotoxicity and a low ecotoxicological footprint. Mode-of-action studies revealed that P1-type nucleoside transporters and adenosine kinase are involved in the uptake and activation, respectively. Analogue 3 represents a potent, advanced lead fitting the preferred target product profile for a broad-spectrum trypanocide regardless of the causative species.
Collapse
Affiliation(s)
- Kayhan Ilbeigi
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - An Matheeussen
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - Rik Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - Mathieu Claes
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - Nick Van Reet
- Protozoology Research Group, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Roel Anthonissen
- Sciensano, SD Chemical and Physical Health Risks, 1050 Brussels, Belgium
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, B-9000 Gent, Belgium
| | - Cai Lin
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, B-9000 Gent, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - Clement Regnault
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, Garscube Campus, University of Glasgow, Glasgow G61 1BD, U.K
| | - Phillip Whitfield
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, Garscube Campus, University of Glasgow, Glasgow G61 1BD, U.K
| | - Rajdeep Roy
- iES Landau, Institute for Environmental Sciences, University of Kaiserslautern-Landau (RPTU), 76829 Landau, Germany
| | - Marzuq A Ungogo
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, U.K
| | - Yann G-J Sterckx
- Laboratory of Medical Biochemistry (LMB), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | - Birgit Mertens
- Sciensano, SD Chemical and Physical Health Risks, 1050 Brussels, Belgium
| | - Mirco Bundschuh
- iES Landau, Institute for Environmental Sciences, University of Kaiserslautern-Landau (RPTU), 76829 Landau, Germany
- Department of Aquatic Sciences and Assessment, Swedish University of Agricultural Sciences, 75007 Uppsala, Sweden
| | - Harry P De Koning
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, U.K
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, B-9000 Gent, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
3
|
Renzi G, Ladu F, Carta F, Supuran CT. The carbonic anhydrase enzymes as new targets for the management of neglected tropical diseases. Arch Pharm (Weinheim) 2025; 358:e2400626. [PMID: 39520343 PMCID: PMC11726158 DOI: 10.1002/ardp.202400626] [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: 08/04/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Diseases caused by protozoan parasites represent a huge challenge to global health care, due to the lack of selective and efficient treatments for the management and spreading of such complex pathologies. The protozoans Trypanosoma cruzi (T. cruzi) and Leishmania spp. are the etiological agents of the so-called neglected tropical diseases (NTDs), that is, Chagas disease (CD) and leishmaniasis, respectively. In such a context, the metalloenzymes carbonic anhydrases (CAs; EC 4.2.1.1) emerged as potential protozoan druggable enzymes, being involved in the parasites' life cycle. Several studies suggested the relevance of the protozoan-expressed CAs as future candidates for the management of NTDs.
Collapse
Affiliation(s)
- Gioele Renzi
- NEUROFARBA DepartmentPharmaceutical and Nutraceutical Section, University of FlorenceSesto FiorentinoItaly
| | - Federico Ladu
- Department of Medicine, Surgery and PharmacyUniversity of SassariSassariItaly
| | - Fabrizio Carta
- NEUROFARBA DepartmentPharmaceutical and Nutraceutical Section, University of FlorenceSesto FiorentinoItaly
| | - Claudiu T. Supuran
- NEUROFARBA DepartmentPharmaceutical and Nutraceutical Section, University of FlorenceSesto FiorentinoItaly
| |
Collapse
|
4
|
Pereira SH, Alves FP, Teixeira SMR. Animal Trypanosomiasis: Challenges and Prospects for New Vaccination Strategies. Microorganisms 2024; 12:2575. [PMID: 39770779 PMCID: PMC11678697 DOI: 10.3390/microorganisms12122575] [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: 11/05/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Animal trypanosomiasis, such as nagana, surra, and dourine, represent a significant challenge to animal health and economic development, especially in tropical and subtropical regions where livestock production is an essential component of a country's economy. Despite advances in the control of human trypanosomiasis, animal diseases caused by several species of trypanosomes remain neglected. The lack of funding for the development of new treatments and vaccines contributes to sustaining the severe economic impacts these diseases have on the farming industry, especially in low-income rural areas. Recent advances in the understanding of the immune processes involved during infection have been essential for the development of new approaches towards disease control including vaccines. These new approaches must be part of integrated control programs, which must also include vector management and the awareness of good veterinary practices. Addressing the challenges posed by the control of animal trypanosomiasis requires collaborative and continuous efforts shared among scientists, governments, and the farming industry, if significant progress is to be made to mitigate the impact of these diseases. In this literature review, we discuss the main challenges for the development of vaccines for animal trypanosomiasis and the research underway, including the prospects for employing new vaccine platforms, such as an mRNA vaccine, vector-based vaccine, and CRISPR-attenuated parasite vaccine.
Collapse
Affiliation(s)
- Samille Henriques Pereira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (S.H.P.); (F.P.A.)
| | - Felipe Paladino Alves
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (S.H.P.); (F.P.A.)
| | - Santuza Maria Ribeiro Teixeira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (S.H.P.); (F.P.A.)
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte 31310-260, MG, Brazil
| |
Collapse
|
5
|
Jooste J, Legoabe LJ, Ilbeigi K, Caljon G, Beteck RM. Hydrazinated geraniol derivatives as potential broad-spectrum antiprotozoal agents. Arch Pharm (Weinheim) 2024; 357:e2400430. [PMID: 38982314 DOI: 10.1002/ardp.202400430] [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: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Geraniol, a primary component of several essential oils, has been associated with broad-spectrum antiprotozoal activities, although moderate to weak. This study primarily concentrated on the synthesis of hydrazinated geraniol derivatives as potential antiprotozoal agents. The synthesised compounds were tested in vitro against different parasitic protozoans of clinical relevance, including Trypanosoma brucei brucei, Trypanosoma brucei rhodesiense, Trypanosoma cruzi and Leishmania infantum. Compounds 6, 8, 13, 14 and 15 demonstrated low micromolar activity against the different parasites. Compounds 8, 13, 14 and 15 had the highest efficacy against Trypanosoma brucei rhodesiense, as indicated by their respective IC50 values of 0.74, 0.56, 1.26 and 1.00 µM. Compounds 6, 14 and 15 displayed the best activity against Trypanosoma brucei brucei, with IC50 values of 1.49, 1.48 and 1.85 µM, respectively. The activity of compounds 6, 14 and 15 also extended to intracellular Trypanosoma cruzi, with IC50 values of 5.14, 6.30 and 4.90 µM, respectively. Compound 6, with an IC50 value of 11.73 µM, and compound 14, with an IC50 value of 8.14 µM, demonstrated some modest antileishmanial activity.
Collapse
Affiliation(s)
- Joelien Jooste
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Kayhan Ilbeigi
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Richard M Beteck
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
6
|
Ogbonna EN, Terrell JR, Paul A, Farahat AA, Poon GMK, Boykin DW, Wilson WD. Single GC base pair recognition by a heterocyclic diamidine: structures, affinities, and dynamics. RSC Adv 2024; 14:29675-29682. [PMID: 39297050 PMCID: PMC11408989 DOI: 10.1039/d4ra05957c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024] Open
Abstract
The recognition of specific genomic arrangements by rationally designed small molecules is fundamental for the expansion of targeted gene expression. Here, we report the first X-ray crystal structures that demonstrate single G (guanine) recognition by a highly selective diamidine (DB2447) in a mixed DNA sequence. The study presents detailed structural information on the mechanism of single G recognition by D2447 and its various interactions in the DNA minor groove. Molecular dynamics and binding studies were used to evaluate the details of our reported structures. The study provides structural insight and resources necessary for understanding single G selection in genomic sequences.
Collapse
Affiliation(s)
- Edwin N Ogbonna
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
| | - J Ross Terrell
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
| | - Ananya Paul
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
| | - Abdelbasset A Farahat
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
- Master of Pharmaceutical Science Program, California North State University Elk Grove CA 95757 USA
| | - Gregory M K Poon
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
| | - David W Boykin
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
| | - W David Wilson
- Department of Chemistry and Centre of Diagnostics and Therapeutics, Georgia State University Atlanta GA 30303-3083 USA +1 404-413-5505 +1 404-413-5503
| |
Collapse
|
7
|
Fernando L, Echesabal-Chen J, Miller M, Powell RR, Bruce T, Paul A, Poudyal N, Saliutama J, Parman K, Paul KS, Stamatikos A. Cholesterol Efflux Decreases TLR4-Target Gene Expression in Cultured Macrophages Exposed to T. brucei Ghosts. Microorganisms 2024; 12:1730. [PMID: 39203572 PMCID: PMC11357207 DOI: 10.3390/microorganisms12081730] [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: 08/13/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Trypanosoma brucei causes African trypanosomiasis in humans. Infection with T. brucei elicits a potent pro-inflammatory immune response within infected human hosts, and this response is thought to at least be partially due to Toll-like receptor (TLR) activation. In response to stimulation by lipopolysaccharide and other pathogen antigens, TLR4 translocates to lipid rafts, which induces the expression of pro-inflammatory genes. However, cholesterol efflux is acknowledged as anti-inflammatory due to promoting lipid raft disruption. In this study, we wanted to assess the impact of T. brucei "ghosts", which are non-viable T. brucei essentially devoid of intracellular contents, in stimulating macrophage TLR4 translocation to lipid rafts, and whether promoting cholesterol efflux in macrophages incubated with T. brucei ghosts attenuates TLR4-target gene expression. When cultured macrophages were exposed to T. brucei ghosts, we observed an increase in lipid raft TLR4 protein content, which suggests certain surface molecules of T. brucei serve as ligands for TLR4. However, pretreating macrophages with cholesterol acceptors before T. brucei ghost exposure decreased lipid raft TLR4 protein content and the expression of pro-inflammatory TLR4-target genes. Taken together, these results imply that macrophage cholesterol efflux weakens pro-inflammatory responses which occur from T. brucei infection via increasing macrophage lipid raft disruption.
Collapse
Affiliation(s)
- Lawrence Fernando
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (L.F.); (J.E.-C.)
| | - Jing Echesabal-Chen
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (L.F.); (J.E.-C.)
| | - Murphy Miller
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA;
| | - Rhonda Reigers Powell
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Terri Bruce
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Apurba Paul
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Nava Poudyal
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Joshua Saliutama
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Kristina Parman
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Kimberly S. Paul
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (L.F.); (J.E.-C.)
| |
Collapse
|
8
|
Sharma A, Sanz-Rodriguez CE, Pollastri MP, Purmal A, Mensa-Wilmot K. Multiparameter ranking of carbazoles for anti-trypanosome lead discovery. FRONTIERS IN DRUG DISCOVERY 2024; 4:1430927. [PMID: 40125275 PMCID: PMC11927960 DOI: 10.3389/fddsv.2024.1430927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The criteria for the progression of hits in the discovery of leads for human African trypanosomiasis (HAT), a neglected disease caused by the microbial eukaryote Trypanosoma brucei, are not standardized. Hits are advanced upon meeting thresholds for drug-like molecules. Following those principles, pharmacokinetics (Cmax and AUC0-6h) and anti-trypanosome characteristics predicted the arrest of T. brucei proliferation in mice by three curaxins. Unexpectedly, while CBL0137 cured HAT in a mouse model, CBL0174 and CBL0187-structural analogs of CBL0137 with similar drug-like properties-failed to control T. brucei division. We here propose an alternative strategy that integrates physicochemical, metabolic, pharmacokinetic, pharmacodynamic, tissue distribution, and trypanocidality parameters into calculating a score for ranking compounds in hit-to-lead campaigns. Data from our studies of curaxins support the feasibility of this goal. Serum dropped the anti-trypanosome potency of CBL0174 and CBL0187 considerably. Delayed trypanocidal concentrations (DTC25 and DTC90) were used to study modes of curaxin actions in trypanosomes. Efficacy of CBL0137 in mice correlated with (i) a high AUC0-6h: DTC90 ratio, (ii) blocking of transferrin endocytosis, and (iii) the inhibition of protein synthesis. Hydroxylation of the carbazole prevented CBL0137 from inhibiting endocytosis of transferrin. The multiparametric score "Curaxin HAT lead efficacy (CHLE)" score was calculated using pharmacokinetic, physicochemical, metabolic, brain exposure, and pharmacodynamic data; CBL0137 was the highest scoring hit. Complementing these observations and predictive of performance of curaxins in mice, CBL0137, but not CBL0174 or CBL0187, was trypanocidal after the exposure of trypanosomes to AUC0-6h amounts of the hits for 6 hours in vitro. We discuss a role for CHLE scores in ranking curaxins for anti-HAT lead discovery. The principles used to develop CHLE scores may be used to calculate new ones for other scaffolds during the discovery of leads for HAT or other infectious diseases.
Collapse
Affiliation(s)
- Amrita Sharma
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, United States
| | - Carlos E. Sanz-Rodriguez
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Michael P. Pollastri
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, United States
| | | | - Kojo Mensa-Wilmot
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, United States
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
9
|
Fytas G, Zoidis G, Drakopoulos A, Taylor MC, Kelly JM, Tsatsaroni A, Tsotinis A. New Lipophilic Hydroxamates as Promising Trypanocidal Agents: Design, Synthesis, SAR, and Conformational Behavior Studies. ACS Med Chem Lett 2024; 15:1041-1048. [PMID: 39015276 PMCID: PMC11247629 DOI: 10.1021/acsmedchemlett.4c00111] [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: 03/06/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 07/18/2024] Open
Abstract
A series of novel hydroxamic acid derivatives was designed and synthesized, and their growth inhibitory activity against bloodstream form Trypanosoma brucei was evaluated. These compounds are based on conformationally constrained, lipophilic, spiro carbocyclic 2,6-diketopiperazine (2,6-DKP) scaffolds and bear a side pharmacophoric functionality that contains an acetohydroxamic acid moiety (CH2CONHOH) linked with the imidic nitrogen atom of the 2,6-DKP ring via an acetamido portion [CH2CON(R), R = H, CH3]. Most of these analogues were active in the midnanomolar to low micromolar range against T. brucei. (S)-Isobutyl- or (S)-benzyl-substitution on the methylene carbon located between the amine nitrogen atom and carbonyl of the 2,6-DKP ring was studied. The effect of the methyl-substitution on the nitrogen atom of the acetamido portion in the side pharmacophoric functionality was also examined. Compounds 22 and 23, bearing an isobutyl- or benzyl-substituent, respectively, and concurrently a methyl-substituent, were found to be the most potent hydroxamates of this series (IC50 = 34 and 53 nM, respectively). Both had promising selectivity over the parasite compared to mammalian cells (SI = 940 and 470, respectively). Moreover, an E/Z conformational behavior study on hydroxamic acid 18 and its methyl-substituted counterpart 21 was undertaken using NMR spectroscopy and theoretical calculations.
Collapse
Affiliation(s)
- George Fytas
- Faculty
of Pharmacy, Department of Pharmaceutical Chemistry, University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Grigoris Zoidis
- Faculty
of Pharmacy, Department of Pharmaceutical Chemistry, University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Antonios Drakopoulos
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, Göteborg SE-412 96, Sweden
| | - Martin C. Taylor
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - John M. Kelly
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Alexandra Tsatsaroni
- Faculty
of Pharmacy, Department of Pharmaceutical Chemistry, University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Andrew Tsotinis
- Faculty
of Pharmacy, Department of Pharmaceutical Chemistry, University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| |
Collapse
|
10
|
Buguet AGC. From pole to pole, life-long research of sleep in extreme environments. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae025. [PMID: 38737795 PMCID: PMC11085838 DOI: 10.1093/sleepadvances/zpae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/25/2024] [Indexed: 05/14/2024]
Abstract
In November 1965, Michel Jouvet accepted me into his laboratory in Lyon as a medical student at a time when sleep research was an adventure. After 4 years of investigations in cats, I obtained my medical doctorate. Being a military physician, I was posted to Antarctica for wintering over and was initiated by Jean Rivolier into the psychology of small isolated human groups. I recorded 180 polysomnographic (PSG) nights in eight of my companions. This was my first contribution to research on human sleep under extreme environments and conditions. I then entered René Hénane's military thermophysiology laboratory, where I analyzed thermal exchanges during human sleep in the heat. Back to the cold, I spent 2 years in Canada and analyzed sleep during the Arctic winter under the direction of Manny W. Radomski, who headed the Defense and Civil Institute of Environmental Medicine and judged my PhD dissertation along with my first two mentors. Throughout my career, I worked in collaboration with Manny Radomski under the auspices of the Franco-Canadian Accord for Defence Research. We studied sleep and exercise, sleep deprivation, and recovery with and without chemical help. He also gave me support during several investigations in Africa. There, I studied normal sleep under various tropical climates (warm and dry in Niger, warm and humid in Côte d'Ivoire and Congo, temperate mid-mountain in Angola). I determined that human African trypanosomiasis, the ravaging sleeping sickness or tsetse disease, is not a hypersomnia, but a disorder of circadian rhythms, notably in the sleep-wake cycle.
Collapse
Affiliation(s)
- Alain G C Buguet
- Invited Scientist (ret), Malaria Research Unit, UMR 5246 CNRS, Claude-Bernard Lyon-1 University, Villeurbanne, France
| |
Collapse
|
11
|
Carrillo AK, Kadayat TM, Hwang JY, Chen Y, Zhu F, Holbrook G, Gillingwater K, Connelly MC, Yang L, Kaiser M, Guy RK. Antitrypanosomal Chloronitrobenzamides. J Med Chem 2024; 67:3437-3447. [PMID: 38363074 DOI: 10.1021/acs.jmedchem.3c01680] [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: 02/17/2024]
Abstract
Human African trypanosomiasis (HAT), a neglected tropical disease caused by Trypanosoma brucei gambiense (Tbg) or Trypanosoma brucei rhodesiense (Tbr), remains a significant public health concern with over 55 million people at risk of infection. Current treatments for HAT face the challenges of poor efficacy, drug resistance, and toxicity. This study presents the synthesis and evaluation of chloronitrobenzamides (CNBs) against Trypanosoma species, identifying previously reported compound 52 as a potent and selective orally bioavailable antitrypanosomal agent. 52 was well tolerated in vivo and demonstrated favorable oral pharmacokinetics, maintaining plasma concentrations surpassing the cellular EC50 for over 24 h and achieving peak brain concentrations exceeding 7 μM in rodents after single oral administration (50 mg/kg). Treatment with 52 significantly extended the lifespan of mice infected with Trypanosoma congolense and T. brucei rhodesiense. These results demonstrate that 52 is a strong antitrypanosomal lead with potential for developing treatments for both human and animal African trypanosomiasis.
Collapse
Affiliation(s)
- Angela K Carrillo
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Tara Man Kadayat
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509, United States
| | - Jong Yeon Hwang
- Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Daejeon, KR 34114, United States
| | - Yizhe Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509, United States
| | - Fangyi Zhu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Gloria Holbrook
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Kirsten Gillingwater
- Department of Medical Parasitology & Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil 4123, Switzerland
| | - Michele C Connelly
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Marcel Kaiser
- Department of Medical Parasitology & Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil 4123, Switzerland
- Faculty of Science, University of Basel, Petersplatz 1, Basel 4003, Switzerland
| | - R Kiplin Guy
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509, United States
| |
Collapse
|
12
|
Mahadevan L, Arya H, Droste A, Schliebs W, Erdmann R, Kalel VC. PEX1 is essential for glycosome biogenesis and trypanosomatid parasite survival. Front Cell Infect Microbiol 2024; 14:1274506. [PMID: 38510966 PMCID: PMC10952002 DOI: 10.3389/fcimb.2024.1274506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/09/2024] [Indexed: 03/22/2024] Open
Abstract
Trypanosomatid parasites are kinetoplastid protists that compartmentalize glycolytic enzymes in unique peroxisome-related organelles called glycosomes. The heterohexameric AAA-ATPase complex of PEX1-PEX6 is anchored to the peroxisomal membrane and functions in the export of matrix protein import receptor PEX5 from the peroxisomal membrane. Defects in PEX1, PEX6 or their membrane anchor causes dysfunction of peroxisomal matrix protein import cycle. In this study, we functionally characterized a putative Trypanosoma PEX1 orthologue by bioinformatic and experimental approaches and show that it is a true PEX1 orthologue. Using yeast two-hybrid analysis, we demonstrate that TbPEX1 can bind to TbPEX6. Endogenously tagged TbPEX1 localizes to glycosomes in the T. brucei parasites. Depletion of PEX1 gene expression by RNA interference causes lethality to the bloodstream form trypanosomes, due to a partial mislocalization of glycosomal enzymes to the cytosol and ATP depletion. TbPEX1 RNAi leads to a selective proteasomal degradation of both matrix protein import receptors TbPEX5 and TbPEX7. Unlike in yeast, PEX1 depletion did not result in an accumulation of ubiquitinated TbPEX5 in trypanosomes. As PEX1 turned out to be essential for trypanosomatid parasites, it could provide a suitable drug target for parasitic diseases. The results also suggest that these parasites possess a highly efficient quality control mechanism that exports the import receptors from glycosomes to the cytosol in the absence of a functional TbPEX1-TbPEX6 complex.
Collapse
Affiliation(s)
| | | | | | | | - Ralf Erdmann
- Department of Systems Biochemistry, Faculty of Medicine, Institute for Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Vishal C. Kalel
- Department of Systems Biochemistry, Faculty of Medicine, Institute for Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
13
|
Seetsi A, N'da DD, Molefe-Nyembe N, Suganuma K, Ramatla T, Thekisoe O. In vitro anti-trypanosomal activity of synthetic nitrofurantoin-triazole hybrids against Trypanosoma species causing human African trypanosomosis. Fundam Clin Pharmacol 2024; 38:72-83. [PMID: 37479675 DOI: 10.1111/fcp.12940] [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: 02/10/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023]
Abstract
Human African trypanosomosis (HAT) which is also known as sleeping sickness is caused by Trypanosoma brucei gambiense that is endemic in western and central Africa and T. b. rhodesiense that is endemic in eastern and southern Africa. Drugs used for treatment against HAT first stage have limited effectiveness, and the second stage drugs have been reported to be toxic, expensive, and have time-consuming administration, and parasitic resistance has developed against these drugs. The aim of this study was to evaluate the anti-trypanosomal activity of nitrofurantoin-triazole hybrids against T. b. gambiense and T. b. rhodesiense parasites in vitro. This study screened 19 synthesized nitrofurantoin-triazole (NFT) hybrids on two strains of human trypanosomes, and cytotoxicity was evaluated on Madin-Darby bovine kidney (MDBK) cells. The findings in this study showed that an increase in the chain length and the number of carbon atoms in some n-alkyl hybrids influenced the increase in anti-trypanosomal activity against T. b. gambiense and T. b. rhodesiense. The short-chain n-alkyl hybrids showed decreased activity compared to the long-chain n-alkyl hybrids, with increased activity against both T. b. gambiense and T. b. rhodesiense. Incorporation of additional electron-donating substituents in some NFT hybrids showed increased anti-trypanosomal activity than to electron-withdrawing substituents in NFT hybrids. All 19 NFT hybrids tested displayed better anti-trypanosomal activity against T. b. gambiense than T. b. rhodesiense. The NFT hybrid no. 16 was among the best performing hybrids against both T. b. gambiense (0.08 ± 0.04 μM) and T. b.rhodesiense (0.11 ± 0.06 μM), and its activity might be influenced by the introduction of fluorine in the para-position on the benzyl ring. Remarkably, the NFT hybrids in this study displayed weak to moderate cytotoxicity on MDBK cells. All of the NFT hybrids in this study had selectivity index values ranging from 18 to greater than 915, meaning that they were up to 10-100 times fold selective in their anti-trypanosomal activity. The synthesized NFT hybrids showed strong selectivity >10 to T. b. gambiense and T. b. rhodesiense, which indicates that they qualify from the initial selection criteria for potential hit drugs.
Collapse
Affiliation(s)
- Anna Seetsi
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - David D N'da
- Centre of Excellence for Pharmaceutical Sciences (PHARMACEN), North-West University, Potchefstroom, South Africa
| | - Nthatisi Molefe-Nyembe
- Department of Zoology and Entomology, University of the Free State, Phuthaditjhaba, South Africa
| | - Keisuke Suganuma
- OIE Reference Laboratory for Surra, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Tsepo Ramatla
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Oriel Thekisoe
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| |
Collapse
|
14
|
Ribeiro R, Costa L, Pinto E, Sousa E, Fernandes C. Therapeutic Potential of Marine-Derived Cyclic Peptides as Antiparasitic Agents. Mar Drugs 2023; 21:609. [PMID: 38132930 PMCID: PMC10745025 DOI: 10.3390/md21120609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/18/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Parasitic diseases still compromise human health. Some of the currently available therapeutic drugs have limitations considering their adverse effects, questionable efficacy, and long treatment, which have encouraged drug resistance. There is an urgent need to find new, safe, effective, and affordable antiparasitic drugs. Marine-derived cyclic peptides have been increasingly screened as candidates for developing new drugs. Therefore, in this review, a systematic analysis of the scientific literature was performed and 25 marine-derived cyclic peptides with antiparasitic activity (1-25) were found. Antimalarial activity is the most reported (51%), followed by antileishmanial (27%) and antitrypanosomal (20%) activities. Some compounds showed promising antiparasitic activity at the nM scale, being active against various parasites. The mechanisms of action and targets for some of the compounds have been investigated, revealing different strategies against parasites.
Collapse
Affiliation(s)
- Ricardo Ribeiro
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (R.R.); (L.C.); (E.S.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal;
| | - Lia Costa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (R.R.); (L.C.); (E.S.)
| | - Eugénia Pinto
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal;
- Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (R.R.); (L.C.); (E.S.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal;
| | - Carla Fernandes
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (R.R.); (L.C.); (E.S.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal;
| |
Collapse
|
15
|
Monti L, Liu LJ, Varricchio C, Lucero B, Alle T, Yang W, Bem-Shalom I, Gilson M, Brunden KR, Brancale A, Caffrey CR, Ballatore C. Structure-Activity Relationships, Tolerability and Efficacy of Microtubule-Active 1,2,4-Triazolo[1,5-a]pyrimidines as Potential Candidates to Treat Human African Trypanosomiasis. ChemMedChem 2023; 18:e202300193. [PMID: 37429821 PMCID: PMC10615688 DOI: 10.1002/cmdc.202300193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/12/2023]
Abstract
Tubulin and microtubules (MTs) are potential protein targets to treat parasitic infections and our previous studies have shown that the triazolopyrimidine (TPD) class of MT-active compounds hold promise as antitrypanosomal agents. MT-targeting TPDs include structurally related but functionally diverse congeners that interact with mammalian tubulin at either one or two distinct interfacial binding sites; namely, the seventh and vinca sites, which are found within or between α,β-tubulin heterodimers, respectively. Evaluation of the activity of 123 TPD congeners against cultured Trypanosoma brucei enabled a robust quantitative structure-activity relationship (QSAR) model and the prioritization of two congeners for in vivo pharmacokinetics (PK), tolerability and efficacy studies. Treatment of T. brucei-infected mice with tolerable doses of TPDs significantly decreased blood parasitemia within 24 h. Further, two once-weekly doses at 10 mg/kg of a candidate TPD significantly extended the survival of infected mice relative to infected animals treated with vehicle. Further optimization of dosing and/or the dosing schedule of these CNS-active TPDs may provide alternative treatments for human African trypanosomiasis.
Collapse
Affiliation(s)
- Ludovica Monti
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
- Present affiliation: Chemistry Department, Molecular Sciences Research Hub, Imperial College London, 82 Wood Lane, W12 0BZ, London, UK
| | - Lawrence J Liu
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Carmine Varricchio
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF103NB, Cardiff, UK
| | - Bobby Lucero
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
- Department of Chemistry & Biochemistry, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Thibault Alle
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Wenqian Yang
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Ido Bem-Shalom
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Michael Gilson
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Kurt R Brunden
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, 3600 Spruce Street, 19104-6323, Philadelphia, PA, USA
| | - Andrea Brancale
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF103NB, Cardiff, UK
- Present affiliation: Vysoká škola chemicko-technologická v Praze, Department of Organic Chemistry, Technická 5, 16628, Prague 6, Czech Republic
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| | - Carlo Ballatore
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, 92093, La Jolla, CA, USA
| |
Collapse
|
16
|
Racané L, Ptiček L, Kostrun S, Raić-Malić S, Taylor MC, Delves M, Alsford S, Olmo F, Francisco AF, Kelly JM. Bis-6-amidino-benzothiazole Derivative that Cures Experimental Stage 1 African Trypanosomiasis with a Single Dose. J Med Chem 2023; 66:13043-13057. [PMID: 37722077 PMCID: PMC10544003 DOI: 10.1021/acs.jmedchem.3c01051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Indexed: 09/20/2023]
Abstract
We designed and synthesized a series of symmetric bis-6-amidino-benzothiazole derivatives with aliphatic central units and evaluated their efficacy against bloodstream forms of the African trypanosome Trypanosoma brucei. Of these, a dicationic benzothiazole compound (9a) exhibited sub-nanomolar in vitro potency with remarkable selectivity over mammalian cells (>26,000-fold). Unsubstituted 5-amidine groups and a cyclohexyl spacer were the crucial determinants of trypanocidal activity. In all cases, mice treated with a single dose of 20 mg kg-1 were cured of stage 1 trypanosomiasis. The compound displayed a favorable in vitro ADME profile, with the exception of low membrane permeability. However, we found evidence that uptake by T. brucei is mediated by endocytosis, a process that results in lysosomal sequestration. The compound was also active in low nanomolar concentrations against cultured asexual forms of the malaria parasite Plasmodium falciparum. Therefore, 9a has exquisite cross-species efficacy and represents a lead compound with considerable therapeutic potential.
Collapse
Affiliation(s)
- Livio Racané
- Department
of Applied Chemistry, Faculty of Textile Technology, University of Zagreb, Prilaz baruna Filipovića 28a, 10000 Zagreb, Croatia
| | - Lucija Ptiček
- Department
of Applied Chemistry, Faculty of Textile Technology, University of Zagreb, Prilaz baruna Filipovića 28a, 10000 Zagreb, Croatia
| | - Sanja Kostrun
- Chemistry
Department, Selvita Ltd., Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia
| | - Silvana Raić-Malić
- Department
of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 20, 10000 Zagreb, Croatia
| | - Martin Craig Taylor
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, WC1E 7HT London, U.K.
| | - Michael Delves
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, WC1E 7HT London, U.K.
| | - Sam Alsford
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, WC1E 7HT London, U.K.
| | - Francisco Olmo
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, WC1E 7HT London, U.K.
| | - Amanda Fortes Francisco
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, WC1E 7HT London, U.K.
| | - John M. Kelly
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, Keppel Street, WC1E 7HT London, U.K.
| |
Collapse
|
17
|
Ogbonna E, Paul A, Farahat AA, Terrell JR, Mineva E, Ogbonna V, Boykin DW, Wilson WD. X-ray Structure Characterization of the Selective Recognition of AT Base Pair Sequences. ACS BIO & MED CHEM AU 2023; 3:335-348. [PMID: 37599788 PMCID: PMC10436263 DOI: 10.1021/acsbiomedchemau.3c00002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 08/22/2023]
Abstract
The rational design of small molecules that target specific DNA sequences is a promising strategy to modulate gene expression. This report focuses on a diamidinobenzimidazole compound, whose selective binding to the minor groove of AT DNA sequences holds broad significance in the molecular recognition of AT-rich human promoter sequences. The objective of this study is to provide a more detailed and systematized understanding, at an atomic level, of the molecular recognition mechanism of different AT-specific sequences by a rationally designed minor groove binder. The specialized method of X-ray crystallography was utilized to investigate how the sequence-dependent recognition properties in general, A-tract, and alternating AT sequences affect the binding of diamidinobenzimidazole in the DNA minor groove. While general and A-tract AT sequences give a narrower minor groove, the alternating AT sequences intrinsically have a wider minor groove which typically constricts upon binding. A strong and direct hydrogen bond between the N-H of the benzimidazole and an H-bond acceptor atom in the minor groove is essential for DNA recognition in all sequences described. In addition, the diamidine compound specifically utilizes an interfacial water molecule for its DNA binding. DNA complexes of AATT and AAAAAA recognition sites show that the diamidine compound can bind in two possible orientations with a preference for water-assisted hydrogen bonding at either cationic end. The complex structures of AAATTT, ATAT, ATATAT, and AAAA are bound in a singular orientation. Analysis of the helical parameters shows a minor groove expansion of about 1 Å across all the nonalternating DNA complexes. The results from this systematic approach will convey a greater understanding of the specific recognition of a diverse array of AT-rich sequences by small molecules and more insight into the design of small molecules with enhanced specificity to AT and mixed DNA sequences.
Collapse
Affiliation(s)
- Edwin
N. Ogbonna
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| | - Ananya Paul
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| | - Abdelbasset A. Farahat
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
- Department
of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Master
of Pharmaceutical Sciences Program, California
North State University, 9700 W Taron Dr., Elk Grove, California 95757, United States
| | - J. Ross Terrell
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| | - Ekaterina Mineva
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| | - Victor Ogbonna
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| | - David W Boykin
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| | - W. David Wilson
- Department
of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303-3083, United States
| |
Collapse
|
18
|
McInturff EL, France SP, Leverett CA, Flick AC, Lindsey EA, Berritt S, Carney DW, DeForest JC, Ding HX, Fink SJ, Gibson TS, Gray K, Hubbell AK, Johnson AM, Liu Y, Mahapatra S, McAlpine IJ, Watson RB, O'Donnell CJ. Synthetic Approaches to the New Drugs Approved During 2021. J Med Chem 2023; 66:10150-10201. [PMID: 37528515 DOI: 10.1021/acs.jmedchem.3c00501] [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: 08/03/2023]
Abstract
Each year, new drugs are introduced to the market, representing structures that have affinity for biological targets implicated in human diseases and conditions. These new chemical entities (NCEs), particularly small molecules and antibody-drug conjugates, provide insight into molecular recognition and serve as potential leads for the design of future medicines. This annual review is part of a continuing series highlighting the most likely process-scale synthetic approaches to 35 NCEs that were first approved anywhere in the world during 2021.
Collapse
Affiliation(s)
- Emma L McInturff
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Scott P France
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Carolyn A Leverett
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andrew C Flick
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Erick A Lindsey
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Simon Berritt
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Daniel W Carney
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jacob C DeForest
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10777 Science Center Drive, San Diego, California 92121, United States
| | - Hong X Ding
- Pharmacodia (Beijing) Co. Ltd., Beijing, 100085, China
| | - Sarah J Fink
- Takeda Pharmaceuticals, 125 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Tony S Gibson
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Kaitlyn Gray
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Aran K Hubbell
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amber M Johnson
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yiyang Liu
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Subham Mahapatra
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Indrawan J McAlpine
- Genesis Therapeutics, 11568 Sorrento Valley Road, Suite 8, San Diego, California 92121, United States
| | - Rebecca B Watson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10777 Science Center Drive, San Diego, California 92121, United States
| | - Christopher J O'Donnell
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
19
|
Jamabo M, Mahlalela M, Edkins AL, Boshoff A. Tackling Sleeping Sickness: Current and Promising Therapeutics and Treatment Strategies. Int J Mol Sci 2023; 24:12529. [PMID: 37569903 PMCID: PMC10420020 DOI: 10.3390/ijms241512529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the extracellular protozoan parasite Trypanosoma brucei, and targeted for eradication by 2030. The COVID-19 pandemic contributed to the lengthening of the proposed time frame for eliminating human African trypanosomiasis as control programs were interrupted. Armed with extensive antigenic variation and the depletion of the B cell population during an infectious cycle, attempts to develop a vaccine have remained unachievable. With the absence of a vaccine, control of the disease has relied heavily on intensive screening measures and the use of drugs. The chemotherapeutics previously available for disease management were plagued by issues such as toxicity, resistance, and difficulty in administration. The approval of the latest and first oral drug, fexinidazole, is a major chemotherapeutic achievement for the treatment of human African trypanosomiasis in the past few decades. Timely and accurate diagnosis is essential for effective treatment, while poor compliance and resistance remain outstanding challenges. Drug discovery is on-going, and herein we review the recent advances in anti-trypanosomal drug discovery, including novel potential drug targets. The numerous challenges associated with disease eradication will also be addressed.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Maduma Mahlalela
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Adrienne L. Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Centre (BioBRU), Rhodes University, Makhanda 6139, South Africa;
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| |
Collapse
|
20
|
Pardali V, Giannakopoulou E, Mpekoulis G, Tsopela V, Panos G, Taylor MC, Kelly JM, Vassilaki N, Zoidis G. Novel Lipophilic Hydroxamates Based on Spirocarbocyclic Hydantoin Scaffolds with Potent Antiviral and Trypanocidal Activity. Pharmaceuticals (Basel) 2023; 16:1046. [PMID: 37513957 PMCID: PMC10385743 DOI: 10.3390/ph16071046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Flaviviridae infections, such as those caused by hepatitis C (HCV) and dengue viruses (DENVs), represent global health risks. Infected people are in danger of developing chronic liver failure or hemorrhagic fever, both of which can be fatal if not treated. The tropical parasites Trypanosoma brucei and Trypanosoma cruzi cause enormous socioeconomic burdens in Sub-Saharan Africa and Latin America. Anti-HCV chemotherapy has severe adverse effects and is expensive, whereas dengue has no clinically authorized treatment. Antiparasitic medicines are often toxic and difficult to administer, and treatment failures are widely reported. There is an urgent need for new chemotherapies. Based on our previous research, we have undertaken structural modification of lead compound V with the goal of producing derivatives with both antiviral and trypanocidal activity. The novel spirocarbocyclic-substituted hydantoin analogs were designed, synthesized, and tested for antiviral activity against three HCV genotypes (1b, 3a, 4a), DENV, yellow fever virus (YFV), and two trypanosome species (T. brucei, T. cruzi). The optimization was successful and led to compounds with significant antiviral and trypanocidal activity and exceptional selectivity. Several modifications were made to further investigate the structure-activity relationships (SARs) and confirm the critical role of lipophilicity and conformational degrees of freedom.
Collapse
Affiliation(s)
- Vasiliki Pardali
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Erofili Giannakopoulou
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - George Mpekoulis
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Vassilina Tsopela
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Georgios Panos
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Niki Vassilaki
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Grigoris Zoidis
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| |
Collapse
|
21
|
Mukherjee A, Hossain Z, Erben E, Ma S, Choi JY, Kim HS. Identification of a small-molecule inhibitor that selectively blocks DNA-binding by Trypanosoma brucei replication protein A1. Nat Commun 2023; 14:4390. [PMID: 37474515 PMCID: PMC10359466 DOI: 10.1038/s41467-023-39839-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Replication Protein A (RPA) is a broadly conserved complex comprised of the RPA1, 2 and 3 subunits. RPA protects the exposed single-stranded DNA (ssDNA) during DNA replication and repair. Using structural modeling, we discover an inhibitor, JC-229, that targets RPA1 in Trypanosoma brucei, the causative parasite of African trypanosomiasis. The inhibitor is highly toxic to T. brucei cells, while mildly toxic to human cells. JC-229 treatment mimics the effects of TbRPA1 depletion, including DNA replication inhibition and DNA damage accumulation. In-vitro ssDNA-binding assays demonstrate that JC-229 inhibits the activity of TbRPA1, but not the human ortholog. Indeed, despite the high sequence identity with T. cruzi and Leishmania RPA1, JC-229 only impacts the ssDNA-binding activity of TbRPA1. Site-directed mutagenesis confirms that the DNA-Binding Domain A (DBD-A) in TbRPA1 contains a JC-229 binding pocket. Residue Serine 105 determines specific binding and inhibition of TbRPA1 but not T. cruzi and Leishmania RPA1. Our data suggest a path toward developing and testing highly specific inhibitors for the treatment of African trypanosomiasis.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Public Health Research Institute, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
| | - Zakir Hossain
- Department of Chemistry and Biochemistry, Queens College, New York, NY, 11367, USA
| | - Esteban Erben
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Provincia de Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Provincia de Buenos Aires, Argentina
| | - Shuai Ma
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA
| | - Jun Yong Choi
- Department of Chemistry and Biochemistry, Queens College, New York, NY, 11367, USA.
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, 10016, USA.
| | - Hee-Sook Kim
- Public Health Research Institute, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA.
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA.
| |
Collapse
|
22
|
Rao SPS, Gould MK, Noeske J, Saldivia M, Jumani RS, Ng PS, René O, Chen YL, Kaiser M, Ritchie R, Francisco AF, Johnson N, Patra D, Cheung H, Deniston C, Schenk AD, Cortopassi WA, Schmidt RS, Wiedemar N, Thomas B, Palkar R, Ghafar NA, Manoharan V, Luu C, Gable JE, Wan KF, Myburgh E, Mottram JC, Barnes W, Walker J, Wartchow C, Aziz N, Osborne C, Wagner J, Sarko C, Kelly JM, Manjunatha UH, Mäser P, Jiricek J, Lakshminarayana SB, Barrett MP, Diagana TT. Cyanotriazoles are selective topoisomerase II poisons that rapidly cure trypanosome infections. Science 2023; 380:1349-1356. [PMID: 37384702 DOI: 10.1126/science.adh0614] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/24/2023] [Indexed: 07/01/2023]
Abstract
Millions who live in Latin America and sub-Saharan Africa are at risk of trypanosomatid infections, which cause Chagas disease and human African trypanosomiasis (HAT). Improved HAT treatments are available, but Chagas disease therapies rely on two nitroheterocycles, which suffer from lengthy drug regimens and safety concerns that cause frequent treatment discontinuation. We performed phenotypic screening against trypanosomes and identified a class of cyanotriazoles (CTs) with potent trypanocidal activity both in vitro and in mouse models of Chagas disease and HAT. Cryo-electron microscopy approaches confirmed that CT compounds acted through selective, irreversible inhibition of trypanosomal topoisomerase II by stabilizing double-stranded DNA:enzyme cleavage complexes. These findings suggest a potential approach toward successful therapeutics for the treatment of Chagas disease.
Collapse
Affiliation(s)
- Srinivasa P S Rao
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
- Novartis Institute for Tropical Diseases, Singapore
| | - Matthew K Gould
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jonas Noeske
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Manuel Saldivia
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Rajiv S Jumani
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Pearly S Ng
- Novartis Institute for Tropical Diseases, Singapore
| | - Olivier René
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Yen-Liang Chen
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
- Novartis Institute for Tropical Diseases, Singapore
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| | - Ryan Ritchie
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Nila Johnson
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
| | - Debjani Patra
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Harry Cheung
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Colin Deniston
- Novartis Institutes for BioMedical Research, San Diego, CA, USA
| | | | | | - Remo S Schmidt
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| | - Natalie Wiedemar
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| | - Bryanna Thomas
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Rima Palkar
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
| | | | | | - Catherine Luu
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Jonathan E Gable
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Kah Fei Wan
- Novartis Institute for Tropical Diseases, Singapore
| | - Elmarie Myburgh
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Jeremy C Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, UK
| | - Whitney Barnes
- Novartis Institutes for BioMedical Research, San Diego, CA, USA
| | - John Walker
- Novartis Institutes for BioMedical Research, San Diego, CA, USA
| | - Charles Wartchow
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Natasha Aziz
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Colin Osborne
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Juergen Wagner
- Novartis Institute for Tropical Diseases, Singapore
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Christopher Sarko
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - John M Kelly
- London School of Hygiene and Tropical Medicine, London, UK
| | - Ujjini H Manjunatha
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
- Novartis Institute for Tropical Diseases, Singapore
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| | - Jan Jiricek
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institute for Tropical Diseases, Singapore
| | - Suresh B Lakshminarayana
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
- Novartis Institute for Tropical Diseases, Singapore
| | - Michael P Barrett
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Thierry T Diagana
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
- Novartis Institute for Tropical Diseases, Singapore
| |
Collapse
|
23
|
Monti L, Di Antonio M. G-Quadruplexes as Key Transcriptional Regulators in Neglected Trypanosomatid Parasites. Chembiochem 2023; 24:e202300265. [PMID: 37146230 PMCID: PMC10946822 DOI: 10.1002/cbic.202300265] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/07/2023]
Abstract
G-quadruplexes (G4s) are nucleic acid secondary structures that have been linked to the functional regulation of eukaryotic organisms. G4s have been extensively characterised in humans and emerging evidence suggests that they might also be biologically relevant for human pathogens. This indicates that G4s might represent a novel class of therapeutic targets for tackling infectious diseases. Bioinformatic studies revealed a high prevalence of putative quadruplex-forming sequences (PQSs) in the genome of protozoans, which highlights their potential roles in regulating vital processes of these parasites, including DNA transcription and replication. In this work, we focus on the neglected trypanosomatid parasites, Trypanosoma and Leishmania spp., which cause debilitating and deadly diseases across the poorest populations worldwide. We review three examples where G4-formation might be key to modulate transcriptional activity in trypanosomatids, providing an overview of experimental approaches that can be used to exploit the regulatory roles and relevance of these structures to fight parasitic infections.
Collapse
Affiliation(s)
- Ludovica Monti
- Chemistry Department, Imperial College LondonMolecular Sciences Research Hub82 Wood LaneW12 0BZLondonUK
| | - Marco Di Antonio
- Chemistry Department, Imperial College LondonMolecular Sciences Research Hub82 Wood LaneW12 0BZLondonUK
- The Francis Crick Institute1 Midland RoadNW1 1ATLondonUK
- The Institute of Chemical BiologyMolecular Sciences Research Hub82 Wood LaneW12 0BZLondonUK
| |
Collapse
|
24
|
Pinheiro KMP, Guinati BGS, Moreira NS, Coltro WKT. Low-Cost Microfluidic Systems for Detection of Neglected Tropical Diseases. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2023; 16:117-138. [PMID: 37068747 DOI: 10.1146/annurev-anchem-091522-024759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Neglected tropical diseases (NTDs) affect tropical and subtropical countries and are caused by viruses, bacteria, protozoa, and helminths. These kinds of diseases spread quickly due to the tropical climate and limited access to clean water, sanitation, and health care, which make exposed people more vulnerable. NTDs are reported to be difficult and inefficient to diagnose. As mentioned, most NTDs occur in countries that are socially vulnerable, and the lack of resources and access to modern laboratories and equipment intensify the difficulty of diagnosis and treatment, leading to an increase in the mortality rate. Portable and low-cost microfluidic systems have been widely applied for clinical diagnosis, offering a promising alternative that can meet the needs for fast, affordable, and reliable diagnostic tests in developing countries. This review provides a critical overview of microfluidic devices that have been reported in the literature for the detection of the most common NTDs over the past 5 years.
Collapse
Affiliation(s)
| | | | - Nikaele S Moreira
- Instituto de Química, Universidade Federal de Goiás, Goiânia, Brazil;
| | - Wendell K T Coltro
- Instituto de Química, Universidade Federal de Goiás, Goiânia, Brazil;
- Instituto Nacional de Ciência e Tecnologia de Bioanalítica, Campinas, Brazil
| |
Collapse
|
25
|
García-Estrada C, Pérez-Pertejo Y, Domínguez-Asenjo B, Holanda VN, Murugesan S, Martínez-Valladares M, Balaña-Fouce R, Reguera RM. Further Investigations of Nitroheterocyclic Compounds as Potential Antikinetoplastid Drug Candidates. Biomolecules 2023; 13:biom13040637. [PMID: 37189384 DOI: 10.3390/biom13040637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Due to the lack of specific vaccines, management of the trypanosomatid-caused neglected tropical diseases (sleeping sickness, Chagas disease and leishmaniasis) relies exclusively on pharmacological treatments. Current drugs against them are scarce, old and exhibit disadvantages, such as adverse effects, parenteral administration, chemical instability and high costs which are often unaffordable for endemic low-income countries. Discoveries of new pharmacological entities for the treatment of these diseases are scarce, since most of the big pharmaceutical companies find this market unattractive. In order to fill the pipeline of compounds and replace existing ones, highly translatable drug screening platforms have been developed in the last two decades. Thousands of molecules have been tested, including nitroheterocyclic compounds, such as benznidazole and nifurtimox, which had already provided potent and effective effects against Chagas disease. More recently, fexinidazole has been added as a new drug against African trypanosomiasis. Despite the success of nitroheterocycles, they had been discarded from drug discovery campaigns due to their mutagenic potential, but now they represent a promising source of inspiration for oral drugs that can replace those currently on the market. The examples provided by the trypanocidal activity of fexinidazole and the promising efficacy of the derivative DNDi-0690 against leishmaniasis seem to open a new window of opportunity for these compounds that were discovered in the 1960s. In this review, we show the current uses of nitroheterocycles and the novel derived molecules that are being synthesized against these neglected diseases.
Collapse
Affiliation(s)
- Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Bárbara Domínguez-Asenjo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Vanderlan Nogueira Holanda
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 333031, India
| | - María Martínez-Valladares
- Instituto de Ganadería de Montaña (IGM), Consejo Superior de Investigaciones Científicas-Universidad de León, Carretera León-Vega de Infanzones, Vega de Infanzones, 24346 León, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
26
|
Rojas-Pirela M, Kemmerling U, Quiñones W, Michels PAM, Rojas V. Antimicrobial Peptides (AMPs): Potential Therapeutic Strategy against Trypanosomiases? Biomolecules 2023; 13:biom13040599. [PMID: 37189347 DOI: 10.3390/biom13040599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Trypanosomiases are a group of tropical diseases that have devastating health and socio-economic effects worldwide. In humans, these diseases are caused by the pathogenic kinetoplastids Trypanosoma brucei, causing African trypanosomiasis or sleeping sickness, and Trypanosoma cruzi, causing American trypanosomiasis or Chagas disease. Currently, these diseases lack effective treatment. This is attributed to the high toxicity and limited trypanocidal activity of registered drugs, as well as resistance development and difficulties in their administration. All this has prompted the search for new compounds that can serve as the basis for the development of treatment of these diseases. Antimicrobial peptides (AMPs) are small peptides synthesized by both prokaryotes and (unicellular and multicellular) eukaryotes, where they fulfill functions related to competition strategy with other organisms and immune defense. These AMPs can bind and induce perturbation in cell membranes, leading to permeation of molecules, alteration of morphology, disruption of cellular homeostasis, and activation of cell death. These peptides have activity against various pathogenic microorganisms, including parasitic protists. Therefore, they are being considered for new therapeutic strategies to treat some parasitic diseases. In this review, we analyze AMPs as therapeutic alternatives for the treatment of trypanosomiases, emphasizing their possible application as possible candidates for the development of future natural anti-trypanosome drugs.
Collapse
|
27
|
Monti L, Liu LJ, Varricchio C, Lucero B, Alle T, Yang W, Bem-Shalom I, Gilson M, Brunden KR, Brancale A, Caffrey CR, Ballatore C. Structure-Activity Relationships, Tolerability and Efficacy of Microtubule-Active 1,2,4-Triazolo[1,5- a ]pyrimidines as Potential Candidates to Treat Human African Trypanosomiasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.11.532093. [PMID: 36945407 PMCID: PMC10028969 DOI: 10.1101/2023.03.11.532093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Tubulin and microtubules (MTs) are potential protein targets to treat parasitic infections and our previous studies have shown that the triazolopyrimidine (TPD) class of MT- active compounds hold promise as antitrypanosomal agents. MT-targeting TPDs include structurally related but functionally diverse congeners that interact with mammalian tubulin at either one or two distinct interfacial binding sites; namely, the seventh and vinca sites, which are found within or between α,β-tubulin heterodimers, respectively. Evaluation of the activity of 123 TPD congeners against cultured Trypanosoma brucei enabled a robust quantitative structure-activity relationship (QSAR) model and the prioritization of two congeners for in vivo pharmacokinetics (PK), tolerability and efficacy studies. Treatment of T. brucei -infected mice with tolerable doses of TPDs 3 and 4 significantly decreased blood parasitemia within 24 h. Further, two once-weekly doses of 4 at 10 mg/kg significantly extended the survival of infected mice relative to infected animals treated with vehicle. Further optimization of dosing and/or the dosing schedule of these CNS-active TPDs may provide alternative treatments for human African trypanosomiasis.
Collapse
|
28
|
Pfarr KM, Krome AK, Al-Obaidi I, Batchelor H, Vaillant M, Hoerauf A, Opoku NO, Kuesel AC. The pipeline for drugs for control and elimination of neglected tropical diseases: 1. Anti-infective drugs for regulatory registration. Parasit Vectors 2023; 16:82. [PMID: 36859332 PMCID: PMC9979492 DOI: 10.1186/s13071-022-05581-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/05/2022] [Indexed: 03/03/2023] Open
Abstract
The World Health Organization 'Ending the neglect to attain the Sustainable Development Goals: A road map for neglected tropical diseases 2021-2030' outlines the targets for control and elimination of neglected tropical diseases (NTDs). New drugs are needed to achieve some of them. We are providing an overview of the pipeline for new anti-infective drugs for regulatory registration and steps to effective use for NTD control and elimination. Considering drugs approved for an NTD by at least one stringent regulatory authority: fexinidazole, included in WHO guidelines for Trypanosoma brucei gambiense African trypanosomiasis, is in development for Chagas disease. Moxidectin, registered in 2018 for treatment of individuals ≥ 12 years old with onchocerciasis, is undergoing studies to extend the indication to 4-11-year-old children and obtain additional data to inform WHO and endemic countries' decisions on moxidectin inclusion in guidelines and policies. Moxidectin is also being evaluated for other NTDs. Considering drugs in at least Phase 2 clinical development, a submission is being prepared for registration of acoziborole as an oral treatment for first and second stage T.b. gambiense African trypanosomiasis. Bedaquiline, registered for tuberculosis, is being evaluated for multibacillary leprosy. Phase 2 studies of emodepside and flubentylosin in O. volvulus-infected individuals are ongoing; studies for Trichuris trichuria and hookworm are planned. A trial of fosravuconazole in Madurella mycetomatis-infected patients is ongoing. JNJ-64281802 is undergoing Phase 2 trials for reducing dengue viral load. Studies are ongoing or planned to evaluate oxantel pamoate for onchocerciasis and soil-transmitted helminths, including Trichuris, and oxfendazole for onchocerciasis, Fasciola hepatica, Taenia solium cysticercosis, Echinococcus granulosus and soil-transmitted helminths, including Trichuris. Additional steps from first registration to effective use for NTD control and elimination include country registrations, possibly additional studies to inform WHO guidelines and country policies, and implementation research to address barriers to effective use of new drugs. Relative to the number of people suffering from NTDs, the pipeline is small. Close collaboration and exchange of experience among all stakeholders developing drugs for NTDs may increase the probability that the current pipeline will translate into new drugs effectively implemented in affected countries.
Collapse
Affiliation(s)
- Kenneth M. Pfarr
- grid.15090.3d0000 0000 8786 803XInstitute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany ,grid.452463.2German Center for Infection Research, Partner Site Bonn-Cologne, Bonn, Germany
| | - Anna K. Krome
- grid.10388.320000 0001 2240 3300Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Bonn, Germany
| | - Issraa Al-Obaidi
- grid.11984.350000000121138138Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Hannah Batchelor
- grid.11984.350000000121138138Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Michel Vaillant
- grid.451012.30000 0004 0621 531XCompetence Center for Methodology and Statistics, Luxembourg Institute of Health, Strassen, Grand Duchy of Luxembourg
| | - Achim Hoerauf
- grid.15090.3d0000 0000 8786 803XInstitute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany ,grid.452463.2German Center for Infection Research, Partner Site Bonn-Cologne, Bonn, Germany
| | - Nicholas O. Opoku
- grid.449729.50000 0004 7707 5975Department of Epidemiology and Biostatistics School of Public Health, University of Health and Allied Sciences, Hohoe, Ghana
| | - Annette C. Kuesel
- UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases (WHO/TDR), World Health Organization, Geneva, Switzerland
| |
Collapse
|
29
|
Urán Landaburu L, Didier Garnham M, Agüero F. Targeting trypanosomes: how chemogenomics and artificial intelligence can guide drug discovery. Biochem Soc Trans 2023; 51:195-206. [PMID: 36606702 DOI: 10.1042/bst20220618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
Trypanosomatids are protozoan parasites that cause human and animal neglected diseases. Despite global efforts, effective treatments are still much needed. Phenotypic screens have provided several chemical leads for drug discovery, but the mechanism of action for many of these chemicals is currently unknown. Recently, chemogenomic screens assessing the susceptibility or resistance of parasites carrying genome-wide modifications started to define the mechanism of action of drugs at large scale. In this review, we discuss how genomics is being used for drug discovery in trypanosomatids, how integration of chemical and genomics data from these and other organisms has guided prioritisations of candidate therapeutic targets and additional chemical starting points, and how these data can fuel the expansion of drug discovery pipelines into the era of artificial intelligence.
Collapse
Affiliation(s)
- Lionel Urán Landaburu
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Mercedes Didier Garnham
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| |
Collapse
|
30
|
Maciver SK, Abdelnasir S, Anwar A, Siddiqui R, Khan NA. Modular nanotheranostic agents for protistan parasitic diseases: Magic bullets with tracers. Mol Biochem Parasitol 2023; 253:111541. [PMID: 36603708 DOI: 10.1016/j.molbiopara.2022.111541] [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: 03/13/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023]
Abstract
Protistan parasitic infections contribute significantly to morbidity and mortality, causing more than 2 billion human infections annually. However, current treatments are often limited; due to ineffective drugs and drug resistance, thus better options are urgently required. In the present context, theranostics agents are those that offer simultaneous detection, diagnosis and even treatment of protistan parasitic diseases. "Nanotheranostics" is the term used to describe such agents, that are around 100 nm or less in size. Anti-parasitic activity of nanoparticles (NPs) has been reported, and many have useful intrinsic imaging properties, but it is perhaps their multifunctional nature that offers the greatest potential. NPs may be used as adapters onto which various subunits with different functions may be attached. These subunits may facilitate targeting parasites, coupled with toxins to eradicate parasites, and probe subunits for detection of particles and/or parasites. The modular nature of nano-platforms promises a "mix and match" approach for the construction of tailored agents by using combinations of these subunits against different protistan parasites. Even though many of the subunits have shown promise alone, these have not yet been put together convincingly enough to form working theranostics against protistan parasites. Although the clinical application of nanotheranostics to protistan parasitic infections in humans requires more research, we conclude that they offer not just a realisation of Paul Ehrlich's long imagined "magic bullet" concept, but potentially are magic bullets combined with tracer bullets.
Collapse
Affiliation(s)
- Sutherland Kester Maciver
- Centre for Discovery Brain Science, Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Scotland, UK
| | - Sumayah Abdelnasir
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya 47500, Selangor, Malaysia
| | - Ayaz Anwar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya 47500, Selangor, Malaysia.
| | - Ruqaiyyah Siddiqui
- College of Arts and Sciences, American University of Sharjah, Sharjah, United Arab Emirates; Department of Medical Biology, Faculty of Medicine, Istinye University, Istanbul 34010, Turkey
| | - Naveed Ahmed Khan
- Department of Medical Biology, Faculty of Medicine, Istinye University, Istanbul 34010, Turkey; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
31
|
Francisco KR, Monti L, Yang W, Park H, Liu LJ, Watkins K, Amarasinghe DK, Nalli M, Roberto Polaquini C, Regasini LO, Eduardo Miller Crotti A, Silvestri R, Guidi Magalhães L, Caffrey CR. Structure-activity relationship of dibenzylideneacetone analogs against the neglected disease pathogen, Trypanosoma brucei. Bioorg Med Chem Lett 2023; 81:129123. [PMID: 36608774 PMCID: PMC10072319 DOI: 10.1016/j.bmcl.2023.129123] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/20/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
Trypanosoma brucei is a protozoan parasite that causes Human African Trypanosomiasis (HAT), a neglected tropical disease (NTD) that is endemic in 36 countries in sub-Saharan Africa. Only a handful drugs are available for treatment, and these have limitations, including toxicity and drug resistance. Using the natural product, curcumin, as a starting point, several curcuminoids and related analogs were evaluated against bloodstream forms of T. b. brucei. A particular subset of dibenzylideneacetone (DBA) compounds exhibited potent in vitro antitrypanosomal activity with sub-micromolar EC50 values. A structure-activity relationship study including 26 DBA analogs was initiated, and several compounds exhibited EC50 values as low as 200 nM. Cytotoxicity counter screens in HEK293 cells identified several compounds having selectivity indices above 10. These data suggest that DBAs offer starting points for a new small molecule therapy of HAT.
Collapse
Affiliation(s)
- Karol R Francisco
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Ludovica Monti
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenqian Yang
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hayoung Park
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lawrence J Liu
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kaitlyn Watkins
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dilini K Amarasinghe
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marianna Nalli
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Carlos Roberto Polaquini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
| | - Luis O Regasini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil
| | - Antônio Eduardo Miller Crotti
- Faculty of Philosophy, Sciences, and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | - Romano Silvestri
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Lizandra Guidi Magalhães
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca, SP 14404-600, Brazil
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
32
|
Abstract
Leishmaniasis (visceral and cutaneous), Chagas disease and human African trypanosomiasis cause substantial death and morbidity, particularly in low- and middle-income countries. Although the situation has improved for human African trypanosomiasis, there remains an urgent need for new medicines to treat leishmaniasis and Chagas disease; the clinical development pipeline is particularly sparse for Chagas disease. In this Review, we describe recent advances in our understanding of the biology of the causative pathogens, particularly from the drug discovery perspective, and we explore the progress that has been made in the development of new drug candidates and the identification of promising molecular targets. We also explore the challenges in developing new clinical candidates and discuss potential solutions to overcome such hurdles.
Collapse
|
33
|
Mabille D, Dirkx L, Thys S, Vermeersch M, Montenye D, Govaerts M, Hendrickx S, Takac P, Van Weyenbergh J, Pintelon I, Delputte P, Maes L, Pérez-Morga D, Timmermans JP, Caljon G. Impact of pulmonary African trypanosomes on the immunology and function of the lung. Nat Commun 2022; 13:7083. [PMID: 36400767 PMCID: PMC9674601 DOI: 10.1038/s41467-022-34757-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
Approximately 20% of sleeping sickness patients exhibit respiratory complications, however, with a largely unknown role of the parasite. Here we show that tsetse fly-transmitted Trypanosoma brucei parasites rapidly and permanently colonize the lungs and occupy the extravascular spaces surrounding the blood vessels of the alveoli and bronchi. They are present as nests of multiplying parasites exhibiting close interactions with collagen and active secretion of extracellular vesicles. The local immune response shows a substantial increase of monocytes, macrophages, dendritic cells and γδ and activated αβ T cells and a later influx of neutrophils. Interestingly, parasite presence results in a significant reduction of B cells, eosinophils and natural killer cells. T. brucei infected mice show no infection-associated pulmonary dysfunction, mirroring the limited pulmonary clinical complications during sleeping sickness. However, the substantial reduction of the various immune cells may render individuals more susceptible to opportunistic infections, as evident by a co-infection experiment with respiratory syncytial virus. Collectively, these observations provide insights into a largely overlooked target organ, and may trigger new diagnostic and supportive therapeutic approaches for sleeping sickness.
Collapse
Affiliation(s)
- Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sofie Thys
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Daniel Montenye
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Matthias Govaerts
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Takac
- Institute of Zoology, Slovak Academy of Sciences, 84506, Bratislava, Slovakia
- Scientica, Ltd., 83106, Bratislava, Slovakia
| | - Johan Van Weyenbergh
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - David Pérez-Morga
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
34
|
Álvarez-Rodríguez A, Jin BK, Radwanska M, Magez S. Recent progress in diagnosis and treatment of Human African Trypanosomiasis has made the elimination of this disease a realistic target by 2030. Front Med (Lausanne) 2022; 9:1037094. [PMID: 36405602 PMCID: PMC9669443 DOI: 10.3389/fmed.2022.1037094] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Human African Trypanosomiasis (HAT) is caused by unicellular flagellated protozoan parasites of the genus Trypanosoma brucei. The subspecies T. b. gambiense is mainly responsible for mostly chronic anthroponotic infections in West- and Central Africa, accounting for roughly 95% of all HAT cases. Trypanosoma b. rhodesiense results in more acute zoonotic infections in East-Africa. Because HAT has a two-stage pathogenesis, treatment depends on clinical assessment of patients and the determination whether or not parasites have crossed the blood brain barrier. Today, ultimate confirmation of parasitemia is still done by microscopy analysis. However, the introduction of diagnostic lateral flow devices has been a major contributor to the recent dramatic drop in T. b. gambiense HAT. Other techniques such as loop mediated isothermal amplification (LAMP) and recombinant polymerase amplification (RPA)-based tests have been published but are still not widely used in the field. Most recently, CRISPR-Cas technology has been proposed to improve the intrinsic diagnostic characteristics of molecular approaches. This will become crucial in the near future, as preventing the resurgence of HAT will be a priority and will require tools with extreme high positive and negative predicted values, as well as excellent sensitivity and specificity. As for treatment, pentamidine and suramin have historically been the drugs of choice for the treatment of blood-stage gambiense-HAT and rhodesiense-HAT, respectively. For treatment of second-stage infections, drugs that pass the blood brain barrier are needed, and melarsoprol has been effectively used for both forms of HAT in the past. However, due to the high occurrence of post-treatment encephalopathy, the drug is not recommended for use in T. b. gambiense HAT. Here, a combination therapy of eflornithine and nifurtimox (NECT) has been the choice of treatment since 2009. As this treatment requires IV perfusion of eflornithine, efforts were launched in 2003 by the drugs for neglected disease initiative (DNDi) to find an oral-only therapy solution, suitable for rural sub-Saharan Africa treatment conditions. In 2019 this resulted in the introduction of fexinidazole, with a treatment regimen suitable for both the blood-stage and non-severe second-stage T. b. gambiense infections. Experimental treatment of T. b. rhodesiense HAT has now been initiated as well.
Collapse
Affiliation(s)
- Andrés Álvarez-Rodríguez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bo-Kyung Jin
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- *Correspondence: Stefan Magez,
| |
Collapse
|
35
|
Knieß R, Leeder W, Reißig P, Geyer FK, Göringer HU. Core-Shell DNA-Cholesterol Nanoparticles Exert Lysosomolytic Activity in African Trypanosomes. Chembiochem 2022; 23:e202200410. [PMID: 36040754 PMCID: PMC9826209 DOI: 10.1002/cbic.202200410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/18/2022] [Indexed: 01/11/2023]
Abstract
Trypanosoma brucei is the causal infectious agent of African trypanosomiasis in humans and Nagana in livestock. Both diseases are currently treated with a small number of chemotherapeutics, which are hampered by a variety of limitations reaching from efficacy and toxicity complications to drug-resistance problems. Here, we explore the forward design of a new class of synthetic trypanocides based on nanostructured, core-shell DNA-lipid particles. In aqueous solution, the particles self-assemble into micelle-type structures consisting of a solvent-exposed, hydrophilic DNA shell and a hydrophobic lipid core. DNA-lipid nanoparticles have membrane-adhesive qualities and can permeabilize lipid membranes. We report the synthesis of DNA-cholesterol nanoparticles, which specifically subvert the membrane integrity of the T. brucei lysosome, killing the parasite with nanomolar potencies. Furthermore, we provide an example of the programmability of the nanoparticles. By functionalizing the DNA shell with a spliced leader (SL)-RNA-specific DNAzyme, we target a second trypanosome-specific pathway (dual-target approach). The DNAzyme provides a backup to counteract the recovery of compromised parasites, which reduces the risk of developing drug resistance.
Collapse
Affiliation(s)
- Robert Knieß
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - Wolf‐Matthias Leeder
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - Paul Reißig
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - Felix Klaus Geyer
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - H. Ulrich Göringer
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| |
Collapse
|
36
|
Li B, Wang K, Yue H, Drichel A, Lin J, Su Z, Rueping M. Catalyst-Free C(sp 2)-H Borylation through Aryl Radical Generation from Thiophenium Salts via Electron Donor-Acceptor Complex Formation. Org Lett 2022; 24:7434-7439. [PMID: 36191259 DOI: 10.1021/acs.orglett.2c03008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aryl borates lie at the heart of carbon-carbon bond couplings, and they are widely applied to the synthesis of functional materials, pharmaceutical compounds, and natural products. Currently, synthetic methods for aryl borates are mostly limited to metal-catalyzed routes, and nonmetallic strategies remain comparatively underdeveloped. Herein, we report a mild, scalable, visible-light-induced cross-coupling between aryl dibenzothiophenium triflate salts and bis(catecholato)-diboron for the construction of C-B bonds in the absence of base, transition metal-ligand complex, or photoredox catalyst. Mechanistic studies reveal that this transformation is achieved through an electron donor-acceptor (EDA) complex activation in the absence of a catalyst. The mild reaction conditions allow the preparation of aromatic borates in good yields with excellent functional group tolerance. This photochemical protocol was also successfully applied to the late-stage modification of natural products and the synthesis of a drug intermediate, greatly demonstrating broadened utility.
Collapse
Affiliation(s)
- Bo Li
- King Abdullah University of Science and Technology (KAUST), KAUST Catalysis Center (KCC), Thuwal23955-6900, Saudi Arabia.,Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074Aachen, Germany
| | - Ke Wang
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074Aachen, Germany
| | - Huifeng Yue
- King Abdullah University of Science and Technology (KAUST), KAUST Catalysis Center (KCC), Thuwal23955-6900, Saudi Arabia
| | - Alwin Drichel
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074Aachen, Germany
| | - Jingjing Lin
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074Aachen, Germany
| | - Zhenying Su
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074Aachen, Germany
| | - Magnus Rueping
- King Abdullah University of Science and Technology (KAUST), KAUST Catalysis Center (KCC), Thuwal23955-6900, Saudi Arabia
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Gambiense human African trypanosomiasis (gHAT), a disease that has killed hundreds of thousands as recently as the 1990s, could be on the verge of elimination or even eradication. This review describes recent developments that give us reasons for optimism as well as some caveats. RECENT FINDINGS New developments in diagnostic and vector control tools, and especially in treatment, make it possible to strive for elimination of transmission of gHAT by 2030, perhaps even eradication. SUMMARY Gambiense human African trypanosomiasis is a deadly infectious disease affecting West and Central Africa, South Sudan and Uganda, and transmitted between humans by tsetse flies. The disease has caused several major epidemics, the latest one in the 1990s. Thanks to recent innovations such as rapid diagnostic tests for population screening, a single-dose oral treatment and a highly efficient vector control strategy, interruption of transmission of the causative parasite is now within reach. If indeed gHAT has an exclusively human reservoir, this could even result in eradication of the disease. Even if there were an animal reservoir, on the basis of epidemiological data, it plays a limited role. Maintaining adequate postelimination surveillance in known historic foci, using the newly developed tools, should be sufficient to prevent any future resurgence.
Collapse
|
38
|
Kasozi KI, MacLeod ET, Welburn SC. Systematic Review and Meta-Analysis on Human African Trypanocide Resistance. Pathogens 2022; 11:pathogens11101100. [PMID: 36297157 PMCID: PMC9612373 DOI: 10.3390/pathogens11101100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background Human African trypanocide resistance (HATr) is a challenge for the eradication of Human African Trypansomiaisis (HAT) following the widespread emergence of increased monotherapy drug treatment failures against Trypanosoma brucei gambiense and T. b. rhodesiense that are associated with changes in pathogen receptors. Methods: Electronic searches of 12 databases and 3 Google search websites for human African trypanocide resistance were performed using a keyword search criterion applied to both laboratory and clinical studies. Fifty-one publications were identified and included in this study using the PRISMA checklist. Data were analyzed using RevMan and random effect sizes were computed for the statistics at the 95% confidence interval. Results: Pentamidine/melarsoprol/nifurtimox cross-resistance is associated with loss of the T. brucei adenosine transporter 1/purine 2 gene (TbAT1/P2), aquaglyceroporins (TbAQP) 2 and 3, followed by the high affinity pentamidine melarsoprol transporter (HAPT) 1. In addition, the loss of the amino acid transporter (AAT) 6 is associated with eflornithine resistance. Nifurtimox/eflornithine combination therapy resistance is associated with AAT6 and nitroreductase loss, and high resistance and parasite regrowth is responsible for treatment relapse. In clinical studies, the TbAT1 proportion of total random effects was 68% (95% CI: 38.0−91.6); I2 = 96.99% (95% CI: 94.6−98.3). Treatment failure rates were highest with melarsoprol followed by eflornithine at 41.49% (95% CI: 24.94−59.09) and 6.56% (3.06−11.25) respectively. HATr-resistant phenotypes used in most laboratory experiments demonstrated significantly higher pentamidine resistance than other trypanocides. Conclusion: The emergence of drug resistance across the spectrum of trypanocidal agents that are used to treat HAT is a major threat to the global WHO target to eliminate HAT by 2030. T. brucei strains were largely resistant to diamidines and the use of high trypanocide concentrations in clinical studies have proved fatal in humans. Studies to develop novel chemotherapeutical agents and identify alternative protein targets could help to reduce the emergence and spread of HATr.
Collapse
Affiliation(s)
- Keneth Iceland Kasozi
- Infection Medicine, Deanery of Biomedical Sciences, Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH8 9JZ, UK
- School of Medicine, Kabale University, Kabale P.O. Box 317, Uganda
- Correspondence: (K.I.K.); (S.C.W.)
| | - Ewan Thomas MacLeod
- Infection Medicine, Deanery of Biomedical Sciences, Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Susan Christina Welburn
- Infection Medicine, Deanery of Biomedical Sciences, Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH8 9JZ, UK
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University, International Campus, 718 East Haizhou Road, Haining 314400, China
- Correspondence: (K.I.K.); (S.C.W.)
| |
Collapse
|
39
|
Ang CW, Lee BM, Jackson CJ, Wang Y, Franzblau SG, Francisco AF, Kelly JM, Bernhardt PV, Tan L, West NP, Sykes ML, Hinton AO, Bolisetti R, Avery VM, Cooper MA, Blaskovich MA. Nitroimidazopyrazinones with Oral Activity against Tuberculosis and Chagas Disease in Mouse Models of Infection. J Med Chem 2022; 65:13125-13142. [DOI: 10.1021/acs.jmedchem.2c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chee Wei Ang
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Science, Monash University Malaysia, Subang Jaya, 47500 Selangor, Malaysia
| | - Brendon M. Lee
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, United States
| | - Colin J. Jackson
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Paul V. Bernhardt
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Melissa L. Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Alexandra O. Hinton
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Raghu Bolisetti
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Matthew A. Cooper
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mark A.T. Blaskovich
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
40
|
Koester DC, Marx VM, Williams S, Jiricek J, Dauphinais M, René O, Miller SL, Zhang L, Patra D, Chen YL, Cheung H, Gable J, Lakshminarayana SB, Osborne C, Galarneau JR, Kulkarni U, Richmond W, Bretz A, Xiao L, Supek F, Wiesmann C, Honnappa S, Be C, Mäser P, Kaiser M, Ritchie R, Barrett MP, Diagana TT, Sarko C, Rao SPS. Discovery of Novel Quinoline-Based Proteasome Inhibitors for Human African Trypanosomiasis (HAT). J Med Chem 2022; 65:11776-11787. [PMID: 35993839 PMCID: PMC9469205 DOI: 10.1021/acs.jmedchem.2c00791] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human African Trypanosomiasis (HAT) is a vector-borne disease caused by kinetoplastid parasites of the Trypanosoma genus. The disease proceeds in two stages, with a hemolymphatic blood stage and a meningo-encephalic brain stage. In the latter stage, the parasite causes irreversible damage to the brain leading to sleep cycle disruption and is fatal if untreated. An orally bioavailable treatment is highly desirable. In this study, we present a brain-penetrant, parasite-selective 20S proteasome inhibitor that was rapidly optimized from an HTS singleton hit to drug candidate compound 7 that showed cure in a stage II mouse efficacy model. Here, we describe hit expansion and lead optimization campaign guided by cryo-electron microscopy and an in silico model to predict the brain-to-plasma partition coefficient Kp as an important parameter to prioritize compounds for synthesis. The model combined with in vitro and in vivo experiments allowed us to advance compounds with favorable unbound brain-to-plasma ratios (Kp,uu) to cure a CNS disease such as HAT.
Collapse
Affiliation(s)
- Dennis C. Koester
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Vanessa M. Marx
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Sarah Williams
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Jan Jiricek
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Maxime Dauphinais
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Olivier René
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Sarah L. Miller
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Lei Zhang
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Debjani Patra
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Yen-Liang Chen
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Harry Cheung
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Jonathan Gable
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Suresh B. Lakshminarayana
- Pharmacokinetic
Sciences, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Colin Osborne
- Pharmacokinetic
Sciences, Pharmacology and Comparative Medicine, Novartis Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Jean-Rene Galarneau
- Preclinical
Safety, Novartis Institutes for Biomedical
Research, Cambridge, Massachusetts 02139, United States
| | - Upendra Kulkarni
- Chemical
and Pharmaceutical Profiling, Novartis Institutes
for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Wendy Richmond
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, San Diego, California 92121, United States
| | - Angela Bretz
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, San Diego, California 92121, United States
| | - Linda Xiao
- Pharmacology, Novartis Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Frantisek Supek
- Novartis
Institutes for Biomedical Research, San Diego, California 92121, United States
| | | | - Srinivas Honnappa
- Novartis
Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Celine Be
- Novartis
Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, CH 4000 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, CH 4000 Basel, Switzerland
| | - Ryan Ritchie
- University of Glasgow, University Place, Glasgow G12 8TA, U.K
| | | | - Thierry T. Diagana
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Christopher Sarko
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Srinivasa P. S. Rao
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| |
Collapse
|
41
|
Mousavi A, Foroumadi P, Emamgholipour Z, Mäser P, Kaiser M, Foroumadi A. 2-(Nitroaryl)-5-Substituted-1,3,4-Thiadiazole Derivatives with Antiprotozoal Activities: In Vitro and In Vivo Study. Molecules 2022; 27:molecules27175559. [PMID: 36080325 PMCID: PMC9457997 DOI: 10.3390/molecules27175559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Nitro-containing compounds are a well-known class of anti-infective agents, especially in the field of anti-parasitic drug discovery. HAT or sleeping sickness is a neglected tropical disease caused by a protozoan parasite, Trypanosoma brucei. Following the approval of fexinidazole as the first oral treatment for both stages of T. b. gambiense HAT, there is an increased interest in developing new nitro-containing compounds against parasitic diseases. In our previous projects, we synthesized several megazole derivatives that presented high activity against Leishmania major promastigotes. Here, we screened and evaluated their trypanocidal activity. Most of the compounds showed submicromolar IC50 against the BSF form of T. b. rhodesiense (STIB 900). To the best of our knowledge, compound 18c is one of the most potent nitro-containing agents reported against HAT in vitro. Compound 18g revealed an acceptable cure rate in the acute mouse model of HAT, accompanied with noteworthy in vitro activity against T. brucei, T. cruzi, and L. donovani. Taken together, these results suggest that these compounds are promising candidates to evaluate their pharmacokinetic and biological profiles in the future.
Collapse
Affiliation(s)
- Alireza Mousavi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Parham Foroumadi
- Department of Medicinal Chemistry, School of Pharmacy, International Campus, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
- Correspondence: (M.K.); (A.F.)
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Correspondence: (M.K.); (A.F.)
| |
Collapse
|
42
|
Nekoei S, Khamesipour F, Habtemariam S, de Souza W, Mohammadi Pour P, Hosseini SR. The anti‐
Trypanosoma
activities of medicinal plants: A systematic review of the literature. Vet Med Sci 2022; 8:2738-2772. [DOI: 10.1002/vms3.912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Shahin Nekoei
- Faculty of Veterinary Medicine Shahrekord Branch Islamic Azad University Shahrekord Iran
| | - Faham Khamesipour
- Faculty of Veterinary Medicine Shahrekord Branch Islamic Azad University Shahrekord Iran
- Center for Research and Training in Skin Diseases and Leprosy Tehran University of Medical Sciences Tehran Iran
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories and Herbal Analysis Services University of Greenwich Central Avenue Chatham‐Maritime Gillingham Kent UK
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho Universidade Federal do Rio de Janeiro Rio de Janeiro RJ Brazil
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens e Centro Nacional de Biologia Estrutural e Bioimagens Universidade Federal do Rio de Janeiro Rio de Janeiro RJ Brazil
| | - Pardis Mohammadi Pour
- Phytochemistry Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Seyed Reza Hosseini
- Faculty of Veterinary Medicine Shahrekord Branch Islamic Azad University Shahrekord Iran
| |
Collapse
|
43
|
Kushwaha V, Capalash N. Aminoacyl-tRNA synthetase (AARS) as an attractive drug target in neglected tropical trypanosomatid diseases-Leishmaniasis, Human African Trypanosomiasis and Chagas disease. Mol Biochem Parasitol 2022; 251:111510. [PMID: 35988745 DOI: 10.1016/j.molbiopara.2022.111510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 10/15/2022]
Abstract
TriTryp diseases (Leishmaniasis, Human African Trypanosomiasis (HAT), and Chagas disease) are devastating parasitic neglected tropical diseases (NTDs) that affect billions of people in developing countries, cause high mortality in humans, and impose a large socio-economic burden. The current treatment options against tritryp diseases are suboptimal and challenging due to the emergence of resistance against available tritryp drugs. Hence, designing and developing effective anti-tritryp drugs with novel targets are required. Aminoacyl-tRNA synthetases (AARSs) involved in specific aminoacylation of transfer RNAs (tRNAs), interrupt protein synthesis through inhibitors, and retard the parasite growth. AaRSs have long been studied as therapeutic targets in bacteria, and three aaRS inhibitors, mupirocin (against IleRS), tavaborole AN2690 (against LeuRS), and halofuginone (against ProRS), are already in clinical practice. The structural differences between tritryp and human aaRSs and the presence of unique sequences (N-terminal domain/C-terminal domain/catalytic domain) make them potential target for developing selective inhibitors. Drugs based on a single aaRS target developed by high-throughput screening (HTS) are less effective due to the emergence of resistance. However, designing multi-targeted drugs may be a better strategy for resistance development. In this perspective, we discuss the characteristics of tritryp aaRSs, sequence conservation in their orthologs and their peculiarities, recent advancements towards the single-target and multi-target aaRS inhibitors developed through rational design.
Collapse
Affiliation(s)
- Vikas Kushwaha
- Department of Biotechnology, Panjab University, Sector-25, South Campus, Chandigarh 160025, India.
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Sector-25, South Campus, Chandigarh 160025, India.
| |
Collapse
|
44
|
Arbon D, Ženíšková K, Šubrtová K, Mach J, Štursa J, Machado M, Zahedifard F, Leštinová T, Hierro-Yap C, Neuzil J, Volf P, Ganter M, Zoltner M, Zíková A, Werner L, Sutak R. Repurposing of MitoTam: Novel Anti-Cancer Drug Candidate Exhibits Potent Activity against Major Protozoan and Fungal Pathogens. Antimicrob Agents Chemother 2022; 66:e0072722. [PMID: 35856666 PMCID: PMC9380531 DOI: 10.1128/aac.00727-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
Many of the currently available anti-parasitic and anti-fungal frontline drugs have severe limitations, including adverse side effects, complex administration, and increasing occurrence of resistance. The discovery and development of new therapeutic agents is a costly and lengthy process. Therefore, repurposing drugs with already established clinical application offers an attractive, fast-track approach for novel treatment options. In this study, we show that the anti-cancer drug candidate MitoTam, a mitochondria-targeted analog of tamoxifen, efficiently eliminates a wide range of evolutionarily distinct pathogens in vitro, including pathogenic fungi, Plasmodium falciparum, and several species of trypanosomatid parasites, causative agents of debilitating neglected tropical diseases. MitoTam treatment was also effective in vivo and significantly reduced parasitemia of two medically important parasites, Leishmania mexicana and Trypanosoma brucei, in their respective animal infection models. Functional analysis in the bloodstream form of T. brucei showed that MitoTam rapidly altered mitochondrial functions, particularly affecting cellular respiration, lowering ATP levels, and dissipating mitochondrial membrane potential. Our data suggest that the mode of action of MitoTam involves disruption of the inner mitochondrial membrane, leading to rapid organelle depolarization and cell death. Altogether, MitoTam is an excellent candidate drug against several important pathogens, for which there are no efficient therapies and for which drug development is not a priority.
Collapse
Affiliation(s)
- Dominik Arbon
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Kateřina Ženíšková
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Karolína Šubrtová
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Mach
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Štursa
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Marta Machado
- Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Farnaz Zahedifard
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Tereza Leštinová
- Faculty of Sciences, Charles University, Department of Parasitology, Prague, Czech Republic
| | - Carolina Hierro-Yap
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Faculty of Sciences, Charles University, Department of Parasitology, Prague, Czech Republic
| | - Markus Ganter
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Zoltner
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Alena Zíková
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lukáš Werner
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Robert Sutak
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
45
|
Venturelli A, Tagliazucchi L, Lima C, Venuti F, Malpezzi G, Magoulas GE, Santarem N, Calogeropoulou T, Cordeiro-da-Silva A, Costi MP. Current Treatments to Control African Trypanosomiasis and One Health Perspective. Microorganisms 2022; 10:microorganisms10071298. [PMID: 35889018 PMCID: PMC9321528 DOI: 10.3390/microorganisms10071298] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Human African Trypanosomiasis (HAT, sleeping sickness) and Animal African Trypanosomiasis (AAT) are neglected tropical diseases generally caused by the same etiological agent, Trypanosoma brucei. Despite important advances in the reduction or disappearance of HAT cases, AAT represents a risky reservoir of the infections. There is a strong need to control AAT, as is claimed by the European Commission in a recent document on the reservation of antimicrobials for human use. Control of AAT is considered part of the One Health approach established by the FAO program against African Trypanosomiasis. Under the umbrella of the One Health concepts, in this work, by analyzing the pharmacological properties of the therapeutic options against Trypanosoma brucei spp., we underline the need for clearer and more defined guidelines in the employment of drugs designed for HAT and AAT. Essential requirements are addressed to meet the challenge of drug use and drug resistance development. This approach shall avoid inter-species cross-resistance phenomena and retain drugs therapeutic activity.
Collapse
Affiliation(s)
- Alberto Venturelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - Lorenzo Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
- Doctorate School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Clara Lima
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Federica Venuti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - Giulia Malpezzi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - George E. Magoulas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (G.E.M.); (T.C.)
| | - Nuno Santarem
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Theodora Calogeropoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (G.E.M.); (T.C.)
| | - Anabela Cordeiro-da-Silva
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
- Correspondence:
| |
Collapse
|
46
|
Zara L, Moraca F, Van Muijlwijk-Koezen JE, Zarzycka B, Abel R, de Esch IJP. Exploring the Activity Profile of TbrPDEB1 and hPDE4 Inhibitors Using Free Energy Perturbation. ACS Med Chem Lett 2022; 13:904-910. [PMID: 35707144 PMCID: PMC9190044 DOI: 10.1021/acsmedchemlett.1c00690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/13/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Lorena Zara
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Francesca Moraca
- Schrodinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Jacqueline E. Van Muijlwijk-Koezen
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Barbara Zarzycka
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Robert Abel
- Schrodinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Iwan J. P. de Esch
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
47
|
Probing Adamantane Arylhydroxamic Acids against Trypanosoma brucei and Trypanosoma cruzi. MOLBANK 2022. [DOI: 10.3390/m1363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In this work, we present the synthesis and the anti-trypanosomal activity of the 2-(4-(adamant-1-yl)phenyl)-N-hydroxyarylamides, 1a,b and the 2-(4-(adamant-1-yl)phenoxy)-N-hydroxyacetamide, 1c. The 4-(adamant-1-yl)phenyl- and 4-(adamant-1-yl)phenoxy- moieties, which are endowed with promising drug-like properties, are functionalized at the side chain termini as hydroxamic acids. The phenoxy acetohydroxamic derivative, 1c, shows the most interesting profile in terms of activity and toxicity against trypanosomes and merits further investigation.
Collapse
|
48
|
Bobba V, Li Y, Afrin M, Dano R, Zhang W, Li B, Su B. Synthesis and biological evaluation of imidamide analogs as selective anti-trypanosomal agents. Bioorg Med Chem 2022; 61:116740. [PMID: 35396128 PMCID: PMC9074797 DOI: 10.1016/j.bmc.2022.116740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/18/2022]
Abstract
Human African trypanosomiasis is caused by a protozoan parasite Trypanosoma brucei majorly infecting people living in sub-Saharan Africa. Current limited available treatments suffer from drug resistance, severe adverse effects, low efficacy, and costly administrative procedures in African countries with limited medical resources. Therefore, there is always a perpetual demand for advanced drug development and invention of new strategies to combat the disease. Previous work in our lab generated a library of sulfonamide analogs as selective tubulin inhibitors, based on the structural difference between mammalian and trypanosome tubulin proteins. Further lead derivatization was performed in the current study and generated 25 potential drug candidates to improve the drug efficacy and uptake by selectively targeting the parasite's P2 membrane transporter protein with imidamide moiety. One of the newly synthesized analogs, compound 25 with a di-imidamide moiety, has shown greater potency with an IC50 of 1 nM to selectively inhibit the growth of trypanosome cells without affecting the viability of mammalian cells. Western blot analyses reveal that the compound suppressed tubulin polymerization in T. brucei cells. A detailed structure-activity relationship (SAR) was summarized that will be used to guide future lead optimization.
Collapse
Affiliation(s)
- Viharika Bobba
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA
| | - Yaxin Li
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA
| | - Marjia Afrin
- Department of Biological, Geological, and Environmental Sciences, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA
| | - Raina Dano
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA
| | - Wenjing Zhang
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA
| | - Bibo Li
- Department of Biological, Geological, and Environmental Sciences, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA.
| | - Bin Su
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH, 44115, USA.
| |
Collapse
|
49
|
Abacha YZ, Forkuo AD, Gbedema SY, Mittal N, Ottilie S, Rocamora F, Winzeler EA, van Schalkwyk DA, Kelly JM, Taylor MC, Reader J, Birkholtz LM, Lisgarten DR, Cockcroft JK, Lisgarten JN, Palmer RA, Talbert RC, Shnyder SD, Wright CW. Semi-Synthetic Analogues of Cryptolepine as a Potential Source of Sustainable Drugs for the Treatment of Malaria, Human African Trypanosomiasis, and Cancer. Front Pharmacol 2022; 13:875647. [PMID: 35600849 PMCID: PMC9119314 DOI: 10.3389/fphar.2022.875647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/08/2022] [Indexed: 12/11/2022] Open
Abstract
The prospect of eradicating malaria continues to be challenging in the face of increasing parasite resistance to antimalarial drugs so that novel antimalarials active against asexual, sexual, and liver-stage malaria parasites are urgently needed. In addition, new antimalarials need to be affordable and available to those most in need and, bearing in mind climate change, should ideally be sustainable. The West African climbing shrub Cryptolepis sanguinolenta is used traditionally for the treatment of malaria; its principal alkaloid, cryptolepine (1), has been shown to have antimalarial properties, and the synthetic analogue 2,7-dibromocryptolepine (2) is of interest as a lead toward new antimalarial agents. Cryptolepine (1) was isolated using a two-step Soxhlet extraction of C. sanguinolenta roots, followed by crystallization (yield 0.8% calculated as a base with respect to the dried roots). Semi-synthetic 7-bromo- (3), 7, 9-dibromo- (4), 7-iodo- (5), and 7, 9-dibromocryptolepine (6) were obtained in excellent yields by reaction of 1 with N-bromo- or N-iodosuccinimide in trifluoroacetic acid as a solvent. All compounds were active against Plasmodia in vitro, but 6 showed the most selective profile with respect to Hep G2 cells: P. falciparum (chloroquine-resistant strain K1), IC50 = 0.25 µM, SI = 113; late stage, gametocytes, IC50 = 2.2 µM, SI = 13; liver stage, P. berghei sporozoites IC50 = 6.13 µM, SI = 4.6. Compounds 3-6 were also active against the emerging zoonotic species P. knowlesi with 5 being the most potent (IC50 = 0.11 µM). In addition, 3-6 potently inhibited T. brucei in vitro at nM concentrations and good selectivity with 6 again being the most selective (IC50 = 59 nM, SI = 478). These compounds were also cytotoxic to wild-type ovarian cancer cells as well as adriamycin-resistant and, except for 5, cisplatin-resistant ovarian cancer cells. In an acute oral toxicity test in mice, 3-6 did not exhibit toxic effects at doses of up to 100 mg/kg/dose × 3 consecutive days. This study demonstrates that C. sanguinolenta may be utilized as a sustainable source of novel compounds that may lead to the development of novel agents for the treatment of malaria, African trypanosomiasis, and cancer.
Collapse
Affiliation(s)
- Yabalu Z. Abacha
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, United Kingdom,Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Maiduguri, Nigeria
| | - Arnold Donkor Forkuo
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Stephen Y. Gbedema
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, KNUST, Kumasi, Ghana
| | - Nimisha Mittal
- Malaria Drug Accelerator (MalDA) Consortium, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Sabine Ottilie
- Malaria Drug Accelerator (MalDA) Consortium, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Frances Rocamora
- Malaria Drug Accelerator (MalDA) Consortium, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Elizabeth A. Winzeler
- Malaria Drug Accelerator (MalDA) Consortium, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Donelly A. van Schalkwyk
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, South Africa
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, South Africa
| | - David R. Lisgarten
- Biomolecular Research Group, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, United Kingdom
| | - Jeremy K. Cockcroft
- Department of Chemistry, Christopher Ingold Laboratories, University College London, London, United Kingdom
| | | | - Rex A. Palmer
- Department of Crystallography, Biochemical Sciences, Birkbeck College, University of London, London, United Kingdom
| | - Rosemary C. Talbert
- Biomolecular Research Group, School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, United Kingdom
| | - Steven D. Shnyder
- School of Pharmacy and Medical Sciences, Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Colin W. Wright
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, United Kingdom,*Correspondence: Colin W. Wright,
| |
Collapse
|
50
|
Idro R, Ogwang R, Barragan A, Raimondo JV, Masocha W. Neuroimmunology of Common Parasitic Infections in Africa. Front Immunol 2022; 13:791488. [PMID: 35222377 PMCID: PMC8866860 DOI: 10.3389/fimmu.2022.791488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
Parasitic infections of the central nervous system are an important cause of morbidity and mortality in Africa. The neurological, cognitive, and psychiatric sequelae of these infections result from a complex interplay between the parasites and the host inflammatory response. Here we review some of the diseases caused by selected parasitic organisms known to infect the nervous system including Plasmodium falciparum, Toxoplasma gondii, Trypanosoma brucei spp., and Taenia solium species. For each parasite, we describe the geographical distribution, prevalence, life cycle, and typical clinical symptoms of infection and pathogenesis. We pay particular attention to how the parasites infect the brain and the interaction between each organism and the host immune system. We describe how an understanding of these processes may guide optimal diagnostic and therapeutic strategies to treat these disorders. Finally, we highlight current gaps in our understanding of disease pathophysiology and call for increased interrogation of these often-neglected disorders of the nervous system.
Collapse
Affiliation(s)
- Richard Idro
- College of Health Sciences, Makerere University, Kampala, Uganda.,Centre of Tropical Neuroscience, Kitgum, Uganda.,Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Rodney Ogwang
- College of Health Sciences, Makerere University, Kampala, Uganda.,Centre of Tropical Neuroscience, Kitgum, Uganda.,Kenya Medical Research Institute (KEMRI) - Wellcome Trust Research Programme, Nairobi, Kenya
| | - Antonio Barragan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Joseph Valentino Raimondo
- Division of Cell Biology, Department of Human Biology, Neuroscience Institute and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Willias Masocha
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat, Kuwait
| |
Collapse
|