1
|
Barrett MP. Transforming the chemotherapy of human African trypanosomiasis. Clin Microbiol Rev 2025; 38:e0015323. [PMID: 39772631 PMCID: PMC11905362 DOI: 10.1128/cmr.00153-23] [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: 01/11/2025] Open
Abstract
SUMMARYPrior to 2019, when the orally available drug fexinidazole began its clinical use, the treatment of human African trypanosomiasis (HAT) was complex and unsatisfactory for many reasons. Two sub-species of the Trypanosoma brucei parasite are responsible for HAT, namely the rhodesiense form found in East and Southern Africa and the gambiense form found in Central and West Africa. Diseases caused by both forms manifest in two stages: stage 1 before and stage 2 after central nervous system involvement. Prior to 2019, different drugs were required for each of the two parasite sub-species at each stage. Gambiense disease required pentamidine or nifurtimox-eflornithine combination therapy, while for rhodesiense disease, suramin or melarsoprol was given for stages 1 and 2, respectively. These drugs all suffered complications including protracted administration regimens involving multiple injections with drug-induced adverse effects common. Today, a single drug, fexinidazole, can be given orally in most cases for both diseases at either stage. Another compound, acoziborole, effective in both stages 1 and 2 gambiense disease with a single dosing is anticipated to become available within a few years. Moreover, the recent engagement of multilateral organizations in seeking other compounds that could be used in HAT therapy has also been successful, and a rich vein of new trypanocides has been discovered. Here, the clinical use, modes of action, and resistance risks for drugs used against HAT are discussed.
Collapse
Affiliation(s)
- Michael P. Barrett
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
2
|
Gupta S, Babu MA, Kumar R, Singh TG, Goel A, Rastogi S, Sharma P, Tyagi Y, Goel KK, Kumar B. Exploring USFDA-Approved Imidazole-Based Small Molecules in Drug Discovery: A Mini Perspective. Chem Biodivers 2025:e202403020. [PMID: 40062971 DOI: 10.1002/cbdv.202403020] [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: 11/21/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
In the present work, we have explored the importance of the imidazole ring and its importance in drug discovery, citing the key approvals in the present decade (2013-2024). The pharmacological attribution for the approved drugs revealed that out of 20 approved drugs, 45% of the approvals were made as anti-infectives, followed by approvals under the category of genetic and metabolic disorders, sexual endocrine disorders, anticancer, and to treat blood pressure, gastrointestinal disorders, and neurological conditions. Most approved drugs were dispensed through solid dosage forms (13) and thus had predominantly oral routes beside others. The metabolism pattern revealed that the drugs undergo metabolism via the involvement of multiple enzymes, where CYP3A4 and CYP3A5 were the core enzymes. The excretion pattern of these drugs revealed that the drugs are majorly excreted via the fecal route. The chemical analysis showed that pyrrolidine/pyrrole was the major heterocycle in the approved drugs, followed by the indole ring in the hybridization. Considering the substitution pattern, most drugs possessed amide, amines, and fluoro group as the functional substitution with the 2,4-substitution pattern seen in most approved drugs. Besides this, the three approved drugs were found to possess chiral centers and exhibit chirality. The article also expanded to cover the synthetic routes and metabolic routes for this versatile ring system and case studies for its utility to serve as bioisostere in drug discovery. Furthermore, this article also presents the receptor-ligand interactions of imidazole-based drugs with various target receptors. The present article is, therefore, put forth to assist medicinal chemists and chemists working in drug discovery of this versatile ring system.
Collapse
Affiliation(s)
- Sonali Gupta
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to be University), Haridwar, India
- Department of Chemistry, Gurukul Kangri (Deemed to be University), Haridwar, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Roshan Kumar
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, India
| | - Thakur Gurjeet Singh
- Centre of Research Impact and Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Anjali Goel
- Department of Chemistry, Gurukul Kangri (Deemed to be University), Haridwar, India
| | - Sameer Rastogi
- School of Pharmacy, Noida International University, Greater Noida, India
| | - Pankaj Sharma
- Smt Tarawati Institute of Biomedical and Allied Sciences, Roorkee, India
| | - Yogita Tyagi
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Kapil Kumar Goel
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to be University), Haridwar, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (Central University), Srinagar, Uttarakhand, India
| |
Collapse
|
3
|
Andrés-Rodríguez J, González-Montero MC, García-Fernández N, Calvo-Álvarez E, Pérez-Pertejo MY, Reguera-Torres RM, Balaña-Fouce R, García-Estrada C. Free Radical Production Induced by Nitroimidazole Compounds Lead to Cell Death in Leishmania infantum Amastigotes. Molecules 2024; 29:4041. [PMID: 39274889 PMCID: PMC11396368 DOI: 10.3390/molecules29174041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Leishmania infantum is the vector-borne trypanosomatid parasite causing visceral leishmaniasis in the Mediterranean basin. This neglected tropical disease is treated with a limited number of obsolete drugs that are not exempt from adverse effects and whose overuse has promoted the emergence of resistant pathogens. In the search for novel antitrypanosomatid molecules that help overcome these drawbacks, drug repurposing has emerged as a good strategy. Nitroaromatic compounds have been found in drug discovery campaigns as promising antileishmanial molecules. Fexinidazole (recently introduced for the treatment of stages 1 and 2 of African trypanosomiasis), and pretomanid, which share the nitroimidazole nitroaromatic structure, have provided antileishmanial activity in different studies. In this work, we have tested the in vitro efficacy of these two nitroimidazoles to validate our 384-well high-throughput screening (HTS) platform consisting of L. infantum parasites emitting the near-infrared fluorescent protein (iRFP) as a biomarker of cell viability. These molecules showed good efficacy in both axenic and intramacrophage amastigotes and were poorly cytotoxic in RAW 264.7 and HepG2 cultures. Fexinidazole and pretomanid induced the production of ROS in axenic amastigotes but were not able to inhibit trypanothione reductase (TryR), thus suggesting that these compounds may target thiol metabolism through a different mechanism of action.
Collapse
Affiliation(s)
- Julia Andrés-Rodríguez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - María-Cristina González-Montero
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Estefanía Calvo-Álvarez
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - María-Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa-María Reguera-Torres
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), 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
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
4
|
Janse van Rensburg HD, Suganuma K, N'Da DD. In vitro trypanocidal activities and structure-activity relationships of ciprofloxacin analogs. Mol Divers 2024; 28:2667-2680. [PMID: 37481633 DOI: 10.1007/s11030-023-10704-9] [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: 03/14/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Tropical diseases, such as African trypanosomiasis, by their nature and prevalence lack the necessary urgency regarding drug development, despite the increasing need for novel, structurally diverse antitrypanosomal drugs, using different mechanisms of action that would improve drug efficacy and safety. Traditionally antibacterial agents, the fluoroquinolones, reportedly possess in vitro trypanocidal activities against Trypanosoma brucei organisms. During our research, the fluroquinolone, ciprofloxacin (1), and its analogs (2-24) were tested against bloodstream forms of T. brucei brucei, T. b. gambiense, T. b. rhodesiense, T. evansi, T. equiperdum, and T. congolense and Madin-Darby bovine kidney cells (cytotoxicity). Ciprofloxacin [CPX (1)] demonstrated selective trypanocidal activity against T. congolense (IC50 7.79 µM; SI 39.6), whereas the CPX derivatives (2-10) showed weak selective activity (25 < IC50 < 65 µM; 2 < SI < 4). Selectivity and activity of the CPX and 1,2,3-triazole (TZ) hybrids (11-24) were governed by their chemical functionality at C-3 (carboxylic acid, or 4-methylpiperazinyl amide) and their electronic effect (electron-donating or electron-withdrawing para-benzyl substituent), respectively. Trypanocidal hits in the micromolar range were identified against bloodstream forms of T. congolense [CPX (1); CPX amide derivatives 18: IC50 8.95 µM; SI 16.84; 22: IC50 5.42 µM; SI 25.2] and against T. brucei rhodesiense (CPX acid derivative 13: IC50 4.51 µM; SI 10.2), demonstrating more selectivity toward trypanosomes than mammalian cells. Hence, the trypanocidal hit compound 22 may be optimized by retaining the 4-methylpiperazine amide functional group (C-3) and the TZ moiety at position N-15 and introducing other electron-withdrawing ortho-, meta-, and/or para-substituents on the aryl ring in an effort to improve the pharmacokinetic properties and increase the trypanocidal activity.
Collapse
Affiliation(s)
| | - Keisuke Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido, 080-8555, Japan.
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, 2520, South Africa
| |
Collapse
|
5
|
Rivas F, Del Mármol C, Scalese G, Pérez Díaz L, Machado I, Blacque O, Salazar F, Coitiño EL, Benítez D, Medeiros A, Comini M, Gambino D. Multifunctional Organometallic Compounds Active against Infective Trypanosomes: Ru(II) Ferrocenyl Derivatives with Two Different Bioactive Ligands. Inorg Chem 2024; 63:11667-11687. [PMID: 38860314 DOI: 10.1021/acs.inorgchem.4c01125] [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: 06/12/2024]
Abstract
Human African trypanosomiasis (HAT, sleeping sickness) and American trypanosomiasis (Chagas disease) are endemic zoonotic diseases caused by genomically related trypanosomatid protozoan parasites (Trypanosoma brucei and Trypanosoma cruzi, respectively). Just a few old drugs are available for their treatment, with most of them sharing poor safety, efficacy, and pharmacokinetic profiles. Only fexinidazole has been recently incorporated into the arsenal for the treatment of HAT. In this work, new multifunctional Ru(II) ferrocenyl compounds were rationally designed as potential agents against these pathogens by including in a single molecule 1,1'-bis(diphenylphosphino)ferrocene (dppf) and two bioactive bidentate ligands: pyridine-2-thiolato-1-oxide ligand (mpo) and polypyridyl ligands (NN). Three [Ru(mpo)(dppf)(NN)](PF6) compounds and their derivatives with chloride as a counterion were synthesized and fully characterized in solid state and solution. They showed in vitro activity on bloodstream T. brucei (EC50 = 31-160 nM) and on T. cruzi trypomastigotes (EC50 = 190-410 nM). Compounds showed the lowest EC50 values on T. brucei when compared to the whole set of metal-based compounds previously developed by us. In addition, several of the Ru compounds showed good selectivity toward the parasites, particularly against the highly proliferative bloodstream form of T. brucei. Interaction with DNA and generation of reactive oxygen species (ROS) were ruled out as potential targets and modes of action of the Ru compounds. Biochemical assays and in silico analysis led to the insight that they are able to inhibit the NADH-dependent fumarate reductase from T. cruzi. One representative hit induced a mild oxidation of low molecular weight thiols in T. brucei. The compounds were stable for at least 72 h in two different media and more lipophilic than both bioactive ligands, mpo and NN. An initial assessment of the therapeutic efficacy of one of the most potent and selective candidates, [Ru(mpo)(dppf)(bipy)]Cl, was performed using a murine infection model of acute African trypanosomiasis. This hit compound lacks acute toxicity when applied to animals in the dose/regimen described, but was unable to control parasite proliferation in vivo, probably because of its rapid clearance or low biodistribution in the extracellular fluids. Future studies should investigate the pharmacokinetics of this compound in vivo and involve further research to gain deeper insight into the mechanism of action of the compounds.
Collapse
Affiliation(s)
- Feriannys Rivas
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Carolina Del Mármol
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Gonzalo Scalese
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Leticia Pérez Díaz
- Sección Genómica Funcional, Facultad de Ciencias, Universidad de la República, 11400 Montevideo, Uruguay
| | - Ignacio Machado
- Área Química Analítica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, CH 8057 Zurich, Switzerland
| | - Fabiana Salazar
- Laboratorio de Química Teórica y Computacional (LQTC), Instituto de Química Biológica, Facultad de Ciencias, and Centro de Investigaciones Biomédicas (CeInBio), Universidad de la República, 11400 Montevideo, Uruguay
| | - E Laura Coitiño
- Laboratorio de Química Teórica y Computacional (LQTC), Instituto de Química Biológica, Facultad de Ciencias, and Centro de Investigaciones Biomédicas (CeInBio), Universidad de la República, 11400 Montevideo, Uruguay
| | - Diego Benítez
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Andrea Medeiros
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Marcelo Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| |
Collapse
|
6
|
Vijayasurya, Gupta S, Shah S, Pappachan A. Drug repurposing for parasitic protozoan diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:23-58. [PMID: 38942539 DOI: 10.1016/bs.pmbts.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Protozoan parasites are major hazards to human health, society, and the economy, especially in equatorial regions of the globe. Parasitic diseases, including leishmaniasis, malaria, and others, contribute towards majority of morbidity and mortality. Around 1.1 million people die from these diseases annually. The lack of licensed vaccinations worsens the worldwide impact of these diseases, highlighting the importance of safe and effective medications for their prevention and treatment. However, the appearance of drug resistance in parasites continuously affects the availability of medications. The demand for novel drugs motivates global antiparasitic drug discovery research, necessitating the implementation of many innovative ways to maintain a continuous supply of promising molecules. Drug repurposing has come out as a compelling tool for drug development, offering a cost-effective and efficient alternative to standard de novo approaches. A thorough examination of drug repositioning candidates revealed that certain drugs may not benefit significantly from their original indications. Still, they may exhibit more pronounced effects in other disorders. Furthermore, certain medications can produce a synergistic effect, resulting in enhanced therapeutic effectiveness when given together. In this chapter, we outline the approaches employed in drug repurposing (sometimes referred to as drug repositioning), propose novel strategies to overcome these hurdles and fully exploit the promise of drug repurposing. We highlight a few major human protozoan diseases and a range of exemplary drugs repurposed for various protozoan infections, providing excellent outcomes for each disease.
Collapse
Affiliation(s)
- Vijayasurya
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - Swadha Gupta
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - Smit Shah
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - Anju Pappachan
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India.
| |
Collapse
|
7
|
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
|
8
|
Pelizaro BI, Batista JCZ, Portapilla GB, das Neves AR, Silva F, Carvalho DB, Shiguemoto CYK, Pessatto LR, Paredes-Gamero EJ, Cardoso IA, Luccas PH, Nonato MC, Lopes NP, Galvão F, Oliveira KMP, Cassemiro NS, Silva DB, Piranda EM, Arruda CCP, de Albuquerque S, Baroni ACM. Design and Synthesis of Novel 3-Nitro-1 H-1,2,4-triazole-1,2,3-triazole-1,4-disubstituted Analogs as Promising Antitrypanosomatid Agents: Evaluation of In Vitro Activity against Chagas Disease and Leishmaniasis. J Med Chem 2024; 67:2584-2601. [PMID: 38305199 DOI: 10.1021/acs.jmedchem.3c01745] [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: 02/03/2024]
Abstract
A series of 28 compounds, 3-nitro-1H-1,2,4-triazole, were synthesized by click-chemistry with diverse substitution patterns using medicinal chemistry approaches, such as bioisosterism, Craig-plot, and the Topliss set with excellent yields. Overall, the analogs demonstrated relevant in vitro antitrypanosomatid activity. Analog 15g (R1 = 4-OCF3-Ph, IC50 = 0.09 μM, SI = >555.5) exhibited an outstanding antichagasic activity (Trypanosoma cruzi, Tulahuen LacZ strain) 68-fold more active than benznidazole (BZN, IC50 = 6.15 μM, SI = >8.13) with relevant selectivity index, and suitable LipE = 5.31. 15g was considered an appropriate substrate for the type I nitro reductases (TcNTR I), contributing to a likely potential mechanism of action for antichagasic activity. Finally, 15g showed nonmutagenic potential against Salmonella typhimurium strains (TA98, TA100, and TA102). Therefore, 3-nitro-1H-1,2,4-triazole 15g is a promising antitrypanosomatid candidate for in vivo studies.
Collapse
Affiliation(s)
- Bruno I Pelizaro
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900, Brazil
| | - Jaqueline C Z Batista
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900,Brazil
| | - Gisele B Portapilla
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo CEP 14040-900, Brazil
| | - Amarith R das Neves
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900, Brazil
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900,Brazil
| | - Fernanda Silva
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900,Brazil
| | - Diego B Carvalho
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900, Brazil
| | - Cristiane Y K Shiguemoto
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900, Brazil
| | - Lucas R Pessatto
- Laboratório de Biologia Molecular (BioMol) e Cultivos Celulares, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, Campo Grande,Mato Grosso do Sul CEP 79070-900 ,Brazil
| | - Edgar J Paredes-Gamero
- Laboratório de Biologia Molecular (BioMol) e Cultivos Celulares, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, Campo Grande,Mato Grosso do Sul CEP 79070-900 ,Brazil
| | - Iara A Cardoso
- Laboratório de Cristalografia de Proteínas, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, Ribeirão Preto, São Paulo CEP 14040-903 ,Brazil
| | - Pedro H Luccas
- Laboratório de Cristalografia de Proteínas, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, Ribeirão Preto, São Paulo CEP 14040-903 ,Brazil
| | - M Cristina Nonato
- Laboratório de Cristalografia de Proteínas, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, Ribeirão Preto, São Paulo CEP 14040-903 ,Brazil
| | - Norberto P Lopes
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos, Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, Ribeirão Preto, São Paulo CEP 14040-903, Brazil
| | - Fernanda Galvão
- Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul CEP 79804-970, Brazil
| | - Kelly M P Oliveira
- Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul CEP 79804-970, Brazil
| | - Nadla S Cassemiro
- Laboratório de Produtos Naturais e Espectrometria de Massas (LAPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande ,Mato Grosso do SulCEP 79070-900, Brazil
| | - Denise B Silva
- Laboratório de Produtos Naturais e Espectrometria de Massas (LAPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande ,Mato Grosso do SulCEP 79070-900, Brazil
| | - Eliane M Piranda
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900,Brazil
| | - Carla C P Arruda
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900,Brazil
| | - Sergio de Albuquerque
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo CEP 14040-900, Brazil
| | - Adriano C M Baroni
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul CEP 79070-900, Brazil
| |
Collapse
|
9
|
Escrig JI, Miyamoto Y, Aznar AD, Eckmann L, Debnath A. Antigiardial and antiamebic activities of fexinidazole and its metabolites: new drug leads for giardiasis and amebiasis. Antimicrob Agents Chemother 2024; 68:e0073123. [PMID: 38063401 PMCID: PMC10777846 DOI: 10.1128/aac.00731-23] [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: 06/02/2023] [Accepted: 11/04/2023] [Indexed: 01/11/2024] Open
Abstract
The intestinal parasites Giardia lamblia and Entamoeba histolytica are major causes of morbidity and mortality associated with diarrheal diseases. Metronidazole is the most common drug used to treat giardiasis and amebiasis. Despite its efficacy, treatment failures in giardiasis occur in up to 5%-40% of cases. Potential resistance of E. histolytica to metronidazole is an increasing concern. Therefore, it is critical to search for more effective drugs to treat giardiasis and amebiasis. We identified antigiardial and antiamebic activities of the rediscovered nitroimidazole compound, fexinidazole, and its sulfone and sulfoxide metabolites. Fexinidazole is equally active against E. histolytica and G. lamblia trophozoites, and both metabolites were 3- to 18-fold more active than the parent drug. Fexinidazole and its metabolites were also active against a metronidazole-resistant strain of G. lamblia. G. lamblia and E. histolytica cell extracts exhibited decreased residual nitroreductase activity when metabolites were used as substrates, indicating nitroreductase may be central to the mechanism of action of fexinidazole. In a cell invasion model, fexinidazole and its metabolites significantly reduced the invasiveness of E. histolytica trophozoites through basement membrane matrix. A q.d. oral dose of fexinidazole and its metabolites at 10 mg/kg for 3 days reduced G. lamblia infection significantly in mice compared to control. The newly discovered antigiardial and antiamebic activities of fexinidazole, combined with its FDA-approval and inclusion in the WHO Model List of Essential Medicines for the treatment of human African trypanosomiasis, offer decreased risk and a shortened development timeline toward clinical use of fexinidazole for treatment of giardiasis or amebiasis.
Collapse
Affiliation(s)
- Jose Ignacio Escrig
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Yukiko Miyamoto
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alejandro Delgado Aznar
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Lars Eckmann
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Anjan Debnath
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
10
|
Janse van Rensburg HD, N'Da DD, Suganuma K. In vitro trypanocidal potency and in vivo treatment efficacy of oligomeric ethylene glycol-tethered nitrofurantoin derivatives. Eur J Pharm Sci 2024; 192:106668. [PMID: 38065268 DOI: 10.1016/j.ejps.2023.106668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/22/2023]
Abstract
African trypanosomiasis is a significant vector-borne disease of humans and animals in the tsetse fly belt of Africa, particularly affecting production animals such as cattle, and thus, hindering food security. Trypanosoma congolense (T. congolense), the causative agent of nagana, is livestock's most virulent trypanosome species. There is currently no vaccine against trypanosomiasis; its treatment relies solely on chemotherapy. However, pathogenic resistance has been established against trypanocidal agents in clinical use. This underscores the need to develop new therapeutics to curb trypanosomiasis. Many nitroheterocyclic drugs or compounds, including nitrofurantoin, possess antiparasitic activities in addition to their clinical use as antibiotics. The current study evaluated the in vitro trypanocidal potency and in vivo treatment efficacy of previously synthesized antileishmanial active oligomeric ethylene glycol derivatives of nitrofurantoin. The trypanocidal potency of analogues 2a-o varied among the trypanosome species; however, T. congolense strain IL3000 was more susceptible to these drug candidates than the other human and animal trypanosomes. The arylated analogues 2k (IC50 0.04 µM; SI >6365) and 2l (IC50 0.06 µM; SI 4133) featuring 4-chlorophenoxy and 4-nitrophenoxy moieties, respectively, were revealed as the most promising antitrypanosomal agents of all analogues against T. congolense strain IL3000 trypomastigotes with nanomolar activities. In a preliminary in vivo study involving T. congolense strain IL3000 infected BALB/c mice, the oral administration of 100 mg/kg/day of 2k caused prolonged survival up to 18 days post-infection relative to the infected but untreated control mice which survived 9 days post-infection. However, no cure was achieved due to its poor solubility in the in vivo testing medium, assumably leading to low oral bioavailability. These results confirm the importance of the physicochemical properties lipophilicity and water solubility in attaining not only in vitro trypanocidal potency but also in vivo treatment efficacy. Future work will focus on the chemical optimization of 2k through the investigation of analogues containing solubilizing groups at certain positions on the core structure to improve solubility in the in vivo testing medium which, in the current investigation, is the biggest stumbling block in successfully treating either animal or human Trypanosoma infections.
Collapse
Affiliation(s)
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Keisuke Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
11
|
Carvalho DB, Costa PAN, Portapilla GB, das Neves AR, Shiguemoto CYK, Pelizaro BI, Silva F, Piranda EM, Arruda CCP, Gaspari PDM, Cardoso IA, Luccas PH, Nonato MC, Lopes NP, de Albuquerque S, Baroni ACM. Design, synthesis and antitrypanosomatid activity of 2-nitroimidazole-3,5-disubstituted isoxazole compounds based on benznidazole. Eur J Med Chem 2023; 260:115451. [PMID: 37573209 DOI: 10.1016/j.ejmech.2023.115451] [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: 11/03/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 08/14/2023]
Abstract
Chagas disease and leishmaniasis are neglected diseases of high priority as a public health problem. Pharmacotherapy is based on the administration of a few drugs, which exhibit hazardous adverse effects and toxicity to the patients. Thus, the search for new antitrypanosomatid drugs is imperative to overcome the limitations of the treatments. In this work, 46 2-nitroimidazole 3,5-disubstituted isoxazole compounds were synthesized in good yields by [3 + 2] cycloaddition reaction between terminal acetylene (propargyl-2-nitroimidazole) and chloro-oximes. The compounds were non-toxic to LLC-MK2 cells. Compounds 30, 35, and 44 showed in vitro antichagasic activity, 15-fold, 12-fold, and 10-fold, respectively, more active than benznidazole (BZN). Compounds 30, 35, 44, 45, 53, and 61 acted as substrates for the TcNTR enzyme, indicating that this might be one of the mechanisms of action involved in their antiparasitic activity. Piperazine series and 4-monosubstituted compounds were potent against T. cruzi parasites. Besides the in vitro activity observed in compound 45, the in vivo assay showed that the compound only reduced the parasitemia levels by the seventh-day post-infection (77%, p > 0.001) compared to the control group. However, 45 significantly reduced the parasite load in cardiac tissue (p < 0.01) 11 days post-infection. Compounds 49, 52, and 54 showed antileishmanial activity against intracellular amastigotes of Leishmania (L.) amazonensis at the same range as amphotericin B. These findings highlight the antitrypanosomatid properties of 2-nitroimidazole 3,5-disubstituted isoxazole compounds and the possibility in using them as antitrypanosomatid agents in further studies.
Collapse
Affiliation(s)
- Diego B Carvalho
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79051-470, Brazil
| | - Pedro A N Costa
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79051-470, Brazil; Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79070-900, Brazil
| | - Gisele B Portapilla
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - Universidade de São Paulo, Ribeirão Preto, São Paulo, CEP 14040-900, Brazil
| | - Amarith R das Neves
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79051-470, Brazil; Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79070-900, Brazil
| | - Cristiane Y K Shiguemoto
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79051-470, Brazil
| | - Bruno I Pelizaro
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79051-470, Brazil
| | - Fernanda Silva
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79070-900, Brazil
| | - Eliane M Piranda
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79070-900, Brazil
| | - Carla C P Arruda
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79070-900, Brazil
| | - Priscyla D M Gaspari
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, CEP 14040-903, Ribeirão Preto, SP, Brazil
| | - Iara A Cardoso
- Laboratório de Cristalografia de Proteínas, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, CEP 14040-903, Ribeirão Preto, SP, Brazil
| | - Pedro H Luccas
- Laboratório de Cristalografia de Proteínas, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, CEP 14040-903, Ribeirão Preto, SP, Brazil
| | - M Cristina Nonato
- Laboratório de Cristalografia de Proteínas, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, CEP 14040-903, Ribeirão Preto, SP, Brazil
| | - Norberto P Lopes
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos, Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n Monte Alegre, Ribeirão Preto, SP, CEP 14040-903, Brazil
| | - Sergio de Albuquerque
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - Universidade de São Paulo, Ribeirão Preto, São Paulo, CEP 14040-900, Brazil
| | - Adriano C M Baroni
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grossso do Sul- UFMS, Campo Grande, Mato Grosso do Sul, CEP 79051-470, Brazil.
| |
Collapse
|
12
|
Janse van Rensburg H, N’Da DD, Suganuma K. In Vitro and In Vivo Trypanocidal Efficacy of Nitrofuryl- and Nitrothienylazines. ACS OMEGA 2023; 8:43088-43098. [PMID: 38024678 PMCID: PMC10652724 DOI: 10.1021/acsomega.3c06508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
African trypanosomiasis is a vector-borne disease of animals and humans in the tsetse fly belt of Africa. Trypanosoma congolense ("nagana") is the most pathogenic trypanosome in livestock and causes high morbidity and mortality rates among cattle. In the absence of effective preventative vaccines, the management of trypanosomiasis relies on chemoprophylaxis and/or -therapy. However, the trypanocides in clinical use exhibit poor oral bioavailability and toxicity, and therapeutic failures occur because of resistant strains. Because nitrofurantoin displayed, in addition to its clinical use, promising antiparasitic activity, the current study was conducted to evaluate the in vitro trypanocidal activity and preliminary in vivo treatment efficacy of previously synthesized nitrofuranylazines. The trypanocidal activity of these nitrofuran derivatives varied among the evaluated trypanosome species; however, T. congolense strain IL3000 was more susceptible than other animal and human trypanosomes. The nitrofurylazines 4a (IC50 0.04 μM; SI > 7761) and 7a (IC50 0.03 μM; SI > 9542) as well as the nitrothienylazine 8b (IC50 0.04 μM; SI 232), with nanomolar IC50 values, were revealed as early antitrypanosomal leads. Although these derivatives showed strong trypanocidal activity in vitro, no in vivo treatment efficacy was observed in T. congolense IL3000 infected mice after both oral and intraperitoneal administration in a preliminary study. This was attributed to the poor solubility of the test compounds in the in vivo testing media. Indeed, a challenge in drug discovery is finding a balance between the physicochemical properties of a drug candidate, particularly lipophilicity and water solubility, and maintaining adequate potency to provide an effective dose. Hence, future chemical modifications may be required to generate lead-like to lead-like nitrofuranylazines that possess optimal physicochemical and pharmacokinetic properties while retaining in vitro and, ultimately, in vivo trypanocidal efficacy.
Collapse
Affiliation(s)
| | - David D. N’Da
- Centre
of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Keisuke Suganuma
- National
Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
13
|
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
|
14
|
Dzinamarira T, Almehmadi M, Alsaiari AA, Allahyani M, Aljuaid A, Alsharif A, Khan A, Kamal M, Rabaan AA, Alfaraj AH, AlShehail BM, Alotaibi N, AlShehail SM, Imran M. Highlights on the Development, Related Patents, and Prospects of Lenacapavir: The First-in-Class HIV-1 Capsid Inhibitor for the Treatment of Multi-Drug-Resistant HIV-1 Infection. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1041. [PMID: 37374245 DOI: 10.3390/medicina59061041] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
The multidrug-resistant (MDR) human immunodeficiency virus 1 (HIV-1) infection is an unmet medical need. HIV-1 capsid plays an important role at different stages of the HIV-1 replication cycle and is an attractive drug target for developing therapies against MDR HIV-1 infection. Lenacapavir (LEN) is the first-in-class HIV-1 capsid inhibitor approved by the USFDA, EMA, and Health Canada for treating MDR HIV-1 infection. This article highlights the development, pharmaceutical aspects, clinical studies, patent literature, and future directions on LEN-based therapies. The literature for this review was collected from PubMed, authentic websites (USFDA, EMA, Health Canada, Gilead, and NIH), and the free patent database (Espacenet, USPTO, and Patent scope). LEN has been developed by Gilead and is marketed as Sunlenca (tablet and subcutaneous injection). The long-acting and patient-compliant LEN demonstrated a low level of drug-related mutations, is active against MDR HIV-1 infection, and does not reveal cross-resistance to other anti-HIV drugs. LEN is also an excellent drug for patients having difficult or limited access to healthcare facilities. The literature has established additive/synergistic effects of combining LEN with rilpivirine, cabotegravir, islatravir, bictegravir, and tenofovir. HIV-1 infection may be accompanied by opportunistic infections such as tuberculosis (TB). The associated diseases make HIV treatment complex and warrant drug interaction studies (drug-drug, drug-food, and drug-disease interaction). Many inventions on different aspects of LEN have been claimed in patent literature. However, there is a great scope for developing more inventions related to the drug combination of LEN with anti-HIV/anti-TB drugs in a single dosage form, new formulations, and methods of treating HIV and TB co-infection. Additional research may provide more LEN-based treatments with favorable pharmacokinetic parameters for MDR HIV-1 infections and associated opportunistic infections such as TB.
Collapse
Affiliation(s)
- Tafadzwa Dzinamarira
- School of Health Systems and Public Health, University of Pretoria, Pretoria 0002, South Africa
- ICAP, Columbia University, Harare P.O. Box 28, Zimbabwe
| | - Mazen Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Ahad Amer Alsaiari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Mamdouh Allahyani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Abdulelah Aljuaid
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Abdulaziz Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Abida Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Amal H Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Bashayer M AlShehail
- Pharmacy Practice Department, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Nouf Alotaibi
- Clinical Pharmacy Department, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Shams M AlShehail
- Internal Medicine Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21487, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
15
|
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
|
16
|
Alshrari AS, Hudu SA, Elmigdadi F, Imran M. The Urgent Threat of Clostridioides difficile Infection: A Glimpse of the Drugs of the Future, with Related Patents and Prospects. Biomedicines 2023; 11:biomedicines11020426. [PMID: 36830964 PMCID: PMC9953237 DOI: 10.3390/biomedicines11020426] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Clostridioides difficile infection (CDI) is an urgent threat and unmet medical need. The current treatments for CDI are not enough to fight the burden of CDI and recurrent CDI (r-CDI). This review aims to highlight the future drugs for CDI and their related patented applications. The non-patent literature was collected from PubMed and various authentic websites of pharmaceutical industries. The patent literature was collected from free patent databases. Many possible drugs of the future for CDI, with diverse mechanisms of action, are in development in the form of microbiota-modulating agents (e.g., ADS024, CP101, RBX2660, RBX7455, SYN-004, SER-109, VE303, DAV132, MET-2, and BB128), small molecules (e.g., ridinilazole, ibezapolstat, CRS3123, DNV3837, MGB-BP-3, alanyl-L-glutamine, and TNP-2198), antibodies (e.g., IM-01 and LMN-201), and non-toxic strains of CD (e.g., NTCD-M3). The development of some therapeutic agents (e.g., DS-2969b, OPS-2071, cadazolid, misoprostol, ramoplanin, KB109, LFF571, and Ramizol) stopped due to failed clinical trials or unknown reasons. The patent literature reveals some important inventions for the existing treatments of CDI and supports the possibility of developing more and better CDI-treatment-based inventions, including patient-compliant dosage forms, targeted drug delivery, drug combinations of anti-CDI drugs possessing diverse mechanisms of action, probiotic and enzymatic supplements, and vaccines. The current pipeline of anti-CDI medications appears promising. However, it will be fascinating to see how many of the cited are successful in gaining approval from drug regulators such as the US FDA and becoming medicines for CDI and r-CDI.
Collapse
Affiliation(s)
- Ahmed S. Alshrari
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Shuaibu Abdullahi Hudu
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa 13110, Jordan
- Correspondence: (S.A.H.); (M.I.)
| | - Fayig Elmigdadi
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa 13110, Jordan
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
- Correspondence: (S.A.H.); (M.I.)
| |
Collapse
|
17
|
Oral Brincidofovir Therapy for Monkeypox Outbreak: A Focused Review on the Therapeutic Potential, Clinical Studies, Patent Literature, and Prospects. Biomedicines 2023; 11:biomedicines11020278. [PMID: 36830816 PMCID: PMC9953536 DOI: 10.3390/biomedicines11020278] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The monkeypox disease (MPX) outbreak of 2022 has been reported in more than one hundred countries and is becoming a global concern. Unfortunately, only a few treatments, such as tecovirimat (TCV), are available against MPX. Brincidofovir (BCV) is a United States Food and Drug Administration (USFDA)-approved antiviral against smallpox. This article reviews the potential of BCV for treating MPX and other Orthopoxvirus (OPXVs) diseases. The literature for this review was collected from PubMed, authentic websites (USFDA, Chimerix), and freely available patent databases (USPTO, Espacenet, and Patentscope). BCV (a lipophilic derivative of cidofovir) has been discovered and developed by Chimerix Incorporation, USA. Besides smallpox, BCV has also been tested clinically for various viral infections (adenovirus, cytomegalovirus, ebola virus, herpes simplex virus, and double-stranded DNA virus). Many health agencies and reports have recommended using BCV for MPX. However, no health agency has yet approved BCV for MPX. Accordingly, the off-label use of BCV is anticipated for MPX and various viral diseases. The patent literature revealed some important antiviral compositions of BCV. The authors believe there is a huge opportunity to create novel, inventive, and patentable BCV-based antiviral therapies (new combinations with existing antivirals) for OPXVs illnesses (MPX, smallpox, cowpox, camelpox, and vaccinia). It is also advised to conduct drug interaction (food, drug, and disease interaction) and drug resistance investigations on BCV while developing its combinations with other medications. The BCV-based drug repurposing options are also open for further exploration. BCV offers a promising opportunity for biosecurity against OPXV-based bioterrorism attacks and to control the MPX outbreak of 2022.
Collapse
|
18
|
Alshammari MK, Fatima W, Alraya RA, Khuzaim Alzahrani A, Kamal M, Alshammari RS, Alshammari SA, Alharbi LM, Alsubaie NS, Alosaimi RB, Asdaq SMB, Imran M. Selenium and COVID-19: A spotlight on the clinical trials, inventive compositions, and patent literature. J Infect Public Health 2022; 15:1225-1233. [PMID: 36265330 PMCID: PMC9529344 DOI: 10.1016/j.jiph.2022.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Selenium is an indispensable trace element for all living organisms. It is an essential structural component of several selenium-dependent enzymes, which support the human body's defense mechanism. Recently, the significance of selenium in preventing/treating COVID-19 has been documented in the literature. This review highlights the clinical studies, compositions, and patent literature on selenium to prevent/treat COVID-19. Selenium exerts its anti-COVID-19 action by reducing oxidative stress, declining the expression of the ACE-2 receptor, lowering the discharge of pro-inflammatory substances, and inhibiting the 3CLPro (main protease) and PLpro enzyme of SARS-CoV-2. The data of clinical studies, inventive compositions, and patent literature revealed that selenium monotherapy and its compositions with other nutritional supplements/drugs (vitamin, iron, zinc, copper, ferulic acid, resveratrol, spirulina, N-acetylcysteine, fish oil, many herbs, doxycycline, azithromycin, curcumin, quercetin, etc.,) might be practical to prevent/treat COVID-19. The studies have also suggested a correlation between COVID-19 and selenium deficiency. This indicates that adequate selenium supplementation may provide promising treatment outcomes in COVID-19 patients. The authors foresee the development and commercialization of Selenium-based compositions and dosage forms (spray, inhalers, control release dosage forms, etc.) to battle COVID-19. We also trust that numerous selenium-based compositions are yet to be explored. Accordingly, there is good scope for scientists to work on developing novel and inventive selenium-based compositions to fight against COVID-19. However, there is also a need to consider the narrow therapeutic window and chemical interaction of selenium before developing selenium-based compositions.
Collapse
Affiliation(s)
| | - Waseem Fatima
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia.
| | - Reem Ahmed Alraya
- Department of Pharmaceutical Care, First Health Cluster in Eastern Province, King Fahad Specialist Hospital, Dammam, Saudi Arabia.
| | - A Khuzaim Alzahrani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia.
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Reem Saud Alshammari
- Department of Pharmaceutical Care, Maternity and Children Hospital, Rafha 76321, Saudi Arabia.
| | - Sarah Ayad Alshammari
- Al-Dawaa Medical Services Company (DMSCO), Eastern Province, Al Khobar, Saudi Arabia.
| | | | - Norah Saad Alsubaie
- Sales Department, SPIMACO Addwaeih, Eastern Region Office, Al-Hofuf 9449, Saudi Arabia.
| | | | | | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia.
| |
Collapse
|
19
|
Bernhard S, Kaiser M, Burri C, Mäser P. Fexinidazole for Human African Trypanosomiasis, the Fruit of a Successful Public-Private Partnership. Diseases 2022; 10:90. [PMID: 36278589 PMCID: PMC9589988 DOI: 10.3390/diseases10040090] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 08/14/2023] Open
Abstract
After 100 years of chemotherapy with impractical and toxic drugs, an oral cure for human African trypanosomiasis (HAT) is available: Fexinidazole. In this case, we review the history of drug discovery for HAT with special emphasis on the discovery, pre-clinical development, and operational challenges of the clinical trials of fexinidazole. The screening of the Drugs for Neglected Diseases initiative (DNDi) HAT-library by the Swiss TPH had singled out fexinidazole, originally developed by Hoechst (now Sanofi), as the most promising of a series of over 800 nitroimidazoles and related molecules. In cell culture, fexinidazole has an IC50 of around 1 µM against Trypanosoma brucei and is more than 100-fold less toxic to mammalian cells. In the mouse model, fexinidazole cures both the first, haemolymphatic, and the second, meningoencephalitic stage of the infection, the latter at 100 mg/kg twice daily for 5 days. In patients, the clinical trials managed by DNDi and supported by Swiss TPH mainly conducted in the Democratic Republic of the Congo demonstrated that oral fexinidazole is safe and effective for use against first- and early second-stage sleeping sickness. Based on the positive opinion issued by the European Medicines Agency in 2018, the WHO has released new interim guidelines for the treatment of HAT including fexinidazole as the new therapy for first-stage and non-severe second-stage sleeping sickness caused by Trypanosoma brucei gambiense (gHAT). This greatly facilitates the diagnosis and treatment algorithm for gHAT, increasing the attainable coverage and paving the way towards the envisaged goal of zero transmission by 2030.
Collapse
Affiliation(s)
- Sonja Bernhard
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Christian Burri
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4002 Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- University of Basel, 4002 Basel, Switzerland
| |
Collapse
|
20
|
Beatriz Vermelho A, Rodrigues GC, Nocentini A, Mansoldo FRP, Supuran CT. Discovery of novel drugs for Chagas disease: is carbonic anhydrase a target for antiprotozoal drugs? Expert Opin Drug Discov 2022; 17:1147-1158. [PMID: 36039500 DOI: 10.1080/17460441.2022.2117295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Carbonic anhydrase (CA) arose significant interest as a potential new target for Chagas disease since its discovery in Trypanosoma cruzi in 2013. Benznidazole and Nifurtimox have been used for Chagas disease treatment for 60 years despite all efforts done for obtaining more efficient treatments, acting in the acute and chronic phases of illness, with fewer side effects and resistance induction. AREAS COVERED We discuss the positive and negative aspects of T. cruzi CA (TcCA) studies as a target for developing new drugs. The current research discoveries and the classes of TcCA inhibitors are reviewed. The sulfonamides and their derivatives are the main inhibitor classes, but hydroxamates and the thiols, were investigated too. These compounds inhibited the growth of the evolutive forms of the parasite. A comparative analysis was done with CAs from other Trypanosomatids and protozoans. EXPERT OPINION The search for new targets and drugs is a significant challenge worldwide, and TcCA is a potential candidate for developing new drugs. Several studied inhibitors were active against Trypanosoma cruzi, but their penetration and toxicity problems emerged. New approaches are in progress to obtain inhibitors with desired properties, allowing further steps such as tests using an adequate animal model and subsequent developments for the preclinical testing.
Collapse
Affiliation(s)
- Alane Beatriz Vermelho
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts, and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giseli Capaci Rodrigues
- UNIGRANRIO - Universidade do Grande Rio Programa de Pós-Graduação em Ensino das Ciências, Rio de Janeiro, Brazil
| | - Alessio Nocentini
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence, Italy
| | - Felipe R P Mansoldo
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts, and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudiu T Supuran
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence, Italy
| |
Collapse
|
21
|
The Therapeutic and Prophylactic Potential of Quercetin against COVID-19: An Outlook on the Clinical Studies, Inventive Compositions, and Patent Literature. Antioxidants (Basel) 2022; 11:antiox11050876. [PMID: 35624740 PMCID: PMC9137692 DOI: 10.3390/antiox11050876] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/24/2022] [Indexed: 12/17/2022] Open
Abstract
Quercetin is a phenolic flavonol compound with established antioxidant, anti-inflammatory, and immuno-stimulant properties. Recent studies demonstrate the potential of quercetin against COVID-19. This article highlighted the prophylactic/therapeutic potential of quercetin against COVID-19 in view of its clinical studies, inventions, and patents. The literature for the subject matter was collected utilizing different databases, including PubMed, Sci-Finder, Espacenet, Patentscope, and USPTO. Clinical studies expose the potential of quercetin monotherapy, and also its combination therapy with other compounds, including zinc, vitamin C, curcumin, vitamin D3, masitinib, hydroxychloroquine, azithromycin, and ivermectin. The patent literature also examines claims that quercetin containing nutraceuticals, pharmaceuticals, and dietary supplements, alone or in combination with other drugs/compounds, including favipiravir, remdesivir, molnupiravir, navitoclax, dasatinib, disulfiram, rucaparib, tamarixin, iota-carrageenan, and various herbal extracts (aloe, poria, rosemary, and sphagnum) has potential for use against COVID-19. The literature reveals that quercetin exhibits anti-COVID-19 activity because of its inhibitory effect on the expression of the human ACE2 receptors and the enzymes of SARS-CoV-2 (MPro, PLPro, and RdRp). The USFDA designated quercetin as a “Generally Recognized as Safe” substance for use in the food and beverage industries. It is also an inexpensive and readily available compound. These facts increase the possibility and foreseeability of making novel and economical drug combinations containing quercetin to prevent/treat COVID-19. Quercetin is an acidic compound and shows metabolic interaction with some antivirals, antibiotics, and anti-inflammatory agents. Therefore, the physicochemical and metabolic drug interactions between quercetin and the combined drugs/compounds must be better understood before developing new compositions.
Collapse
|
22
|
Durão R, Ramalhete C, Madureira AM, Mendes E, Duarte N. Plant Terpenoids as Hit Compounds against Trypanosomiasis. Pharmaceuticals (Basel) 2022; 15:ph15030340. [PMID: 35337138 PMCID: PMC8951850 DOI: 10.3390/ph15030340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
Human African trypanosomiasis (sleeping sickness) and American trypanosomiasis (Chagas disease) are vector-borne neglected tropical diseases, caused by the protozoan parasites Trypanosoma brucei and Trypanosoma cruzi, respectively. These diseases were circumscribed to South American and African countries in the past. However, human migration, military interventions, and climate changes have had an important effect on their worldwide propagation, particularly Chagas disease. Currently, the treatment of trypanosomiasis is not ideal, becoming a challenge in poor populations with limited resources. Exploring natural products from higher plants remains a valuable approach to find new hits and enlarge the pipeline of new drugs against protozoal human infections. This review covers the recent studies (2016–2021) on plant terpenoids, and their semi-synthetic derivatives, which have shown promising in vitro and in vivo activities against Trypanosoma parasites.
Collapse
Affiliation(s)
- Raquel Durão
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
| | - Cátia Ramalhete
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
- ATLANTICA—Instituto Universitário, Fábrica da Pólvora de Barcarena, 2730-036 Barcarena, Portugal
| | - Ana Margarida Madureira
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
| | - Eduarda Mendes
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
| | - Noélia Duarte
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
- Correspondence:
| |
Collapse
|