1
|
Nwoke EA, Lowe S, Aldabbagh F, Kalesh K, Kadri H. Nucleoside Analogues for Chagas Disease and Leishmaniasis Therapy: Current Status and Future Perspectives. Molecules 2024; 29:5234. [PMID: 39598623 PMCID: PMC11596272 DOI: 10.3390/molecules29225234] [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: 10/10/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Chagas disease and leishmaniasis are two neglected tropical diseases that affect millions of people in low- and middle-income tropical countries. These diseases caused by protozoan parasites pose significant global health challenges, which have been exacerbated by the recent COVID-19 pandemic. There is an urgent need for novel therapeutics as current treatments are limited by toxicity and drug resistance. Nucleoside analogues, which have been extensively studied and successfully applied in antiviral and antitumor therapies, hold potential that has yet to be fully explored for treating these neglected diseases. In this review, we discuss the use of nucleoside analogues as promising therapeutic agents for Chagas disease and leishmaniasis. After briefly examining the pathology, progression, and current treatment options for these diseases, we provide a comprehensive analysis of the status of nucleoside analogues and explore their prospects. By outlining the current landscape and future directions, this review aims to guide research and development efforts towards more effective nucleoside-based treatments for Chagas disease and leishmaniasis.
Collapse
Affiliation(s)
- Emmanuel Awucha Nwoke
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| | - Silvester Lowe
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| | - Fawaz Aldabbagh
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| | - Karunakaran Kalesh
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK;
| | - Hachemi Kadri
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| |
Collapse
|
2
|
Xie R, Xu J, Shi H, Xiao C, Wang N, Huang N, Yao H. Stereocontrolled Synthesis of Aryl C-Nucleosides under Ambient Conditions. Org Lett 2024; 26:5162-5166. [PMID: 38832704 DOI: 10.1021/acs.orglett.4c01664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
A stereocontrolled synthesis of an aryl C-nucleoside has been developed using D-ribals and arylboronic acids catalyzed by palladium without additional ligands in common solvents under an open-air atmosphere at room temperature. This protocol features very mild conditions, simplicity in operation, exclusive β-stereoselectivity, broad substrate scopes, and good compatibility with reactive amino and hydroxyl groups. The functionalization of unsaturated C-nucleosides and the late-stage glycosylation of natural products/drugs demonstrated the high practicality of this strategy.
Collapse
Affiliation(s)
- Rui Xie
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
| | - Jing Xu
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
| | - Haolin Shi
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
| | - Chenyu Xiao
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
| | - Nengzhong Wang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
- Hubei Three Gorges Laboratory, Yichang 443007, P. R. China
| | - Nianyu Huang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
- Hubei Three Gorges Laboratory, Yichang 443007, P. R. China
| | - Hui Yao
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, P. R. China
- Hubei Three Gorges Laboratory, Yichang 443007, P. R. China
| |
Collapse
|
3
|
Trenaman A, Tinti M, Atrih A, Horn D. Genome-wide screens connect HD82 loss-of-function to purine analog resistance in African trypanosomes. mSphere 2024; 9:e0036323. [PMID: 38126788 PMCID: PMC10826343 DOI: 10.1128/msphere.00363-23] [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: 06/29/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023] Open
Abstract
Nucleoside analogs have been used extensively as anti-infective agents, particularly against viral infections, and have long been considered promising anti-parasitic agents. These pro-drugs are metabolized by host-cell, viral, or parasite enzymes prior to incorporation into DNA, thereby inhibiting DNA replication. Here, we report genes that sensitize African trypanosomes to nucleoside analogs, including the guanosine analog, ganciclovir. We applied ganciclovir selective pressure to a trypanosome genome-wide knockdown library, which yielded nucleoside mono- and diphosphate kinases as hits, validating the approach. The two most dominant hits to emerge, however, were Tb927.6.2800 and Tb927.6.2900, which both encode nuclear proteins; the latter of which is HD82, a SAMHD1-related protein and a putative dNTP triphosphohydrolase. We independently confirmed that HD82, which is conserved among the trypanosomatids, can sensitize Trypanosoma brucei to ganciclovir. Since ganciclovir activity depends upon phosphorylation by ectopically expressed viral thymidine kinase, we also tested the adenosine analog, ara-A, that may be fully phosphorylated by native T. brucei kinase(s). Both Tb927.6.2800 and HD82 knockdowns were resistant to this analog. Tb927.6.2800 knockdown increased sensitivity to hydroxyurea, while dNTP analysis indicated that HD82 is indeed a triphosphohydrolase with dATP as the preferred substrate. Our results provide insights into nucleoside/nucleotide metabolism and nucleoside analog metabolism and resistance in trypanosomatids. We suggest that the product of 6.2800 sensitizes cells to purine analogs through DNA repair, while HD82 does so by reducing the native purine pool.IMPORTANCEThere is substantial interest in developing nucleoside analogs as anti-parasitic agents. We used genome-scale genetic screening and discovered two proteins linked to purine analog resistance in African trypanosomes. Our screens also identified two nucleoside kinases required for pro-drug activation, further validating the approach. The top novel hit, HD82, is related to SAMHD1, a mammalian nuclear viral restriction factor. We validated HD82 and localized the protein to the trypanosome nucleus. HD82 appears to sensitize trypanosomes to nucleoside analogs by reducing native pools of nucleotides, providing insights into both nucleoside/nucleotide metabolism and nucleoside analog resistance in trypanosomatids.
Collapse
Affiliation(s)
- Anna Trenaman
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Michele Tinti
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Abdelmadjid Atrih
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - David Horn
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
4
|
Liu ZQ. Is it still worth renewing nucleoside anticancer drugs nowadays? Eur J Med Chem 2024; 264:115987. [PMID: 38056297 DOI: 10.1016/j.ejmech.2023.115987] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/11/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023]
Abstract
Nucleoside has situated the convergence point in the discovery of novel drugs for decades, and a large number of nucleoside derivatives have been constructed for screening novel pharmacological properties at various experimental platforms. Notably, nearly 20 nucleosides are approved to be used in the clinic treatment of various cancers. Nevertheless, the blossom of synthetic nucleoside analogs in comparison with the scarcity of nucleoside anticancer drugs leads to a question: Is it still worth insisting on the screening of novel anticancer drugs from nucleoside derivatives? Hence, this review attempts to emphasize the importance of nucleoside analogs in the discovery of novel anticancer drugs. Firstly, we introduce the metabolic procedures of nucleoside anticancer drug (such as 5-fluorouracil) and summarize the designing of novel nucleoside anticancer candidates based on clinically used nucleoside anticancer drugs (such as gemcitabine). Furthermore, we collect anticancer properties of some recently synthesized nucleoside analogs, aiming at emphasizing the availability of nucleoside analogs in the discovery of anticancer drugs. Finally, a variety of synthetic strategies including the linkage of sugar moiety with nucleobase scaffold, modifications on the sugar moiety, and variations on the nucleobase structure are collected to exhibit the abundant protocols in the achievement of nucleoside analogs. Taken the above discussions collectively, nucleoside still advantages for the finding of novel anticancer drugs because of the clearly metabolic procedures, successfully clinic applications, and abundantly synthetic routines.
Collapse
Affiliation(s)
- Zai-Qun Liu
- Department of Organic Chemistry, College of Chemistry, Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
5
|
Pal R, Teli G, Akhtar MJ, Matada GSP. Synthetic product-based approach toward potential antileishmanial drug development. Eur J Med Chem 2024; 263:115927. [PMID: 37976706 DOI: 10.1016/j.ejmech.2023.115927] [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: 08/19/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Leishmaniasis is a parasitic disease and is categorized as a tropically neglected disease (NTD) with no effective vaccines available. The available chemotherapeutics against leishmaniasis are associated with an increase in the incidence of toxicity and drug resistance. Consequently, targeting metabolic pathways and enzymes of parasites which differs from the mammalian host can be exploited to treat and overcome the resistance. The classical methods of identifying the structural fragments and the moieties responsible for the biological activities from the standard compounds and their modification are options for developing more effective novel compounds. Significant progress has been made in refining the development of potent non-toxic molecules and addressing the limitations of the current treatment available. Several examples of synthetic product-based approach utilizing their core heterocyclic rings including furan, pyrrole, thiazole, imidazole, pyrazole, triazole, quinazoline, quinoline, pyrimidine, coumarin, indole, acridine, oxadiazole, purine, chalcone, carboline, phenanthrene and metal containing derivatives and their structure-activity relationships are discussed in this review. It also analyses the groups/fragments interacting with the host cell receptors and will support the medicinal chemists with novel antileishmanial agents.
Collapse
Affiliation(s)
- Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Ghanshyam Teli
- School of Pharmacy, Sangam University, Atoon, Bhilwara, 311001, Rajasthan, India
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PO 620, PC 130, Azaiba Bousher, Muscat, Sultanate of Oman
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| |
Collapse
|
6
|
Cardoza S, Shrivash MK, Riva L, Chatterjee AK, Mandal A, Tandon V. Multistep Synthesis of Analogues of Remdesivir: Incorporating Heterocycles at the C-1' Position. J Org Chem 2023; 88:9105-9122. [PMID: 37276453 DOI: 10.1021/acs.joc.3c00754] [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: 06/07/2023]
Abstract
Studies suggest that the 1'β-CN moiety in remdesivir sterically clashes with the Ser861 residue of the RNA-dependent-RNA polymerase (RdRp), causing a delayed chain termination in the RNA replication process. Replacing C1'β-CN with 5-membered heterocycles such as tetrazoles, oxadiazoles, and triazoles can augment the inhibitory activity and pharmacokinetic profile of C-nucleotides. Synthesis of tetrazole-, triazole-, and oxadiazole-integrated C1' analogues of remdesivir was attempted using general synthetic routes. The final compounds 26, 28, and 29 did not inhibit viral replication; however, the synthetic intermediates, i.e., 27 and 50, exhibited an IC90 = 14.1 μM each. The trifluoromethyl-substituted 1,2,4-oxadiazole 59 showed an IC90 of 33.5 μM. This work adds to the growing evidence of the beneficial medicinal impact of C1,1'-disubstituted C-nucleotides.
Collapse
Affiliation(s)
- Savio Cardoza
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Manoj Kumar Shrivash
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad 211012, Uttar Pradesh, India
| | - Laura Riva
- Calibr, Scripps Research, La Jolla, 11119 North Torrey Pines Road Suite 100, California 92037, United States
| | - Arnab K Chatterjee
- Calibr, Scripps Research, La Jolla, 11119 North Torrey Pines Road Suite 100, California 92037, United States
| | - Ajay Mandal
- Symbol Discovery Ltd, ASPIRE-TBI, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad 500046, India
| | - Vibha Tandon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
7
|
Sparrow K, Shrestha R, Wood JM, Clinch K, Hurst BL, Wang H, Gowen BB, Julander JG, Tarbet EB, McSweeney AM, Ward VK, Evans GB, Harris LD. An Isomer of Galidesivir That Potently Inhibits Influenza Viruses and Members of the Bunyavirales Order. ACS Med Chem Lett 2023; 14:506-513. [PMID: 37077387 PMCID: PMC10108398 DOI: 10.1021/acsmedchemlett.3c00048] [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: 02/14/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
We report for the first time the antiviral activities of two iminovirs (antiviral imino-C-nucleosides) 1 and 2, structurally related to galidesivir (Immucillin A, BCX4430). An iminovir containing the 4-aminopyrrolo[2,1-f][1,2,4-triazine] nucleobase found in remdesivir exhibited submicromolar inhibition of multiple strains of influenza A and B viruses, as well as members of the Bunyavirales order. We also report the first syntheses of ProTide prodrugs of iminovir monophosphates, which unexpectedly displayed poorer viral inhibition than their parent nucleosides in vitro. An efficient synthesis of the 4-aminopyrrolo[2,1-f][1,2,4-triazine]-containing iminovir 2 was developed to enable preliminary in vivo studies, wherein it displayed significant toxicity in BALB/c mice and limited protection against influenza. Further modification of this anti-influenza iminovir will therefore be required to improve its therapeutic value.
Collapse
Affiliation(s)
- Kevin
J. Sparrow
- Ferrier
Research Institute, Victoria University
of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand
| | - Rinu Shrestha
- Ferrier
Research Institute, Victoria University
of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - James M. Wood
- Ferrier
Research Institute, Victoria University
of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - Keith Clinch
- Ferrier
Research Institute, Victoria University
of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand
| | - Brett L. Hurst
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322-5600, United States
| | - Hong Wang
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322-5600, United States
| | - Brian B. Gowen
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322-5600, United States
| | - Justin G. Julander
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322-5600, United States
| | - E. Bart Tarbet
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322-5600, United States
| | - Alice M. McSweeney
- Department
of Microbiology & Immunology, University
of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Vernon K. Ward
- The
Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
- Department
of Microbiology & Immunology, University
of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Gary B. Evans
- Ferrier
Research Institute, Victoria University
of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - Lawrence. D. Harris
- Ferrier
Research Institute, Victoria University
of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
8
|
Majumder N, Banerjee A, Saha S. A review on new natural and synthetic anti-leishmanial chemotherapeutic agents and current perspective of treatment approaches. Acta Trop 2023; 240:106846. [PMID: 36720335 DOI: 10.1016/j.actatropica.2023.106846] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
Leishmaniases are considered among the most neglected yet dangerous parasitic diseases worldwide. According to the recent WHO report (Weekly Epidemiological Record, Sep, 2021), 200 countries and territories reported leishmanises cases in 2020; of which 89 (45%) for CL, and 79 (40%) for VL were endemic. Indian subcontinent (India, Bangladesh and Nepal), one of the three eco-epidemiological hotspots of VL, currently reported 18% of the total cases of VL worldwide. Eastern Mediterranean region and the Region of the Americas together reported >90% of the new CL cases, of which >80% were from Afghanistan, Algeria, Brazil, Colombia, Iraq, Pakistan and the Syrian Arab Republic. While considering the current therapeutic options, conventional anti-leishmanial drugs have long been proved to be toxic and/or expensive and have resulted in extensive drug resistance in India. Recent searches for novel anti-leishmanial drugs have led to find out the prime cellular targets and metabolic pathways to bridge the gap between the known facts and unexplored data. Cutting edge knowledge based drug designing has simplified the search for novel molecules with leishmanicidal efficacy by identifying ligand-receptor interactions and has accelerated the cost effective primary discovery of molecules through computational validation against Leishmaniases. This review focuses on the limitations of conventional drugs, and discusses the chemotherapeutic potential of many novel natural and synthetic anti-leishmanial agents reported since the last decade. It is also interpreted that some of the reported molecules might be tested singly or as a part of combinatorial therapy on pre-clinical and clinical level.
Collapse
Affiliation(s)
- Nilanjana Majumder
- Department of Biotechnology, Visva-Bharati, Santiniketan, 731235 West Bengal, India
| | - Antara Banerjee
- Department of Zoology, Bangabasi College, 19 Rajkumar Chakraborty Sarani, Kolkata, 700009 West Bengal, India
| | - Samiran Saha
- Department of Biotechnology, Visva-Bharati, Santiniketan, 731235 West Bengal, India.
| |
Collapse
|
9
|
Barnadas-Carceller B, Martinez-Peinado N, Gómez LC, Ros-Lucas A, Gabaldón-Figueira JC, Diaz-Mochon JJ, Gascon J, Molina IJ, Pineda de las Infantas y Villatoro MJ, Alonso-Padilla J. Identification of compounds with activity against Trypanosoma cruzi within a collection of synthetic nucleoside analogs. Front Cell Infect Microbiol 2023; 12:1067461. [PMID: 36710960 PMCID: PMC9880260 DOI: 10.3389/fcimb.2022.1067461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 01/14/2023] Open
Abstract
Introduction Chagas disease is caused by the protozoan parasite Trypanosoma cruzi, and it is the most important neglected tropical disease in the Americas. Two drugs are available to treat the infection, but their efficacy in the chronic stage of the disease, when most cases are diagnosed, is reduced. Their tolerability is also hindered by common adverse effects, making the development of safer and efficacious alternatives a pressing need. T. cruzi is unable to synthesize purines de novo, relying on a purine salvage pathway to acquire these from its host, making it an attractive target for the development of new drugs. Methods We evaluated the anti-parasitic activity of 23 purine analogs with different substitutions in the complementary chains of their purine rings. We sequentially screened the compounds' capacity to inhibit parasite growth, their toxicity in Vero and HepG2 cells, and their specific capacity to inhibit the development of amastigotes. We then used in-silico docking to identify their likely targets. Results Eight compounds showed specific anti-parasitic activity, with IC50 values ranging from 2.42 to 8.16 μM. Adenine phosphoribosyl transferase, and hypoxanthine-guanine phosphoribosyl transferase, are their most likely targets. Discussion Our results illustrate the potential role of the purine salvage pathway as a target route for the development of alternative treatments against T. cruzi infection, highlithing the apparent importance of specific substitutions, like the presence of benzene groups in the C8 position of the purine ring, consistently associated with a high and specific anti-parasitic activity.
Collapse
Affiliation(s)
- Berta Barnadas-Carceller
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,Secció de Parasitologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Laura Córdoba Gómez
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | | | - Juan J. Diaz-Mochon
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Granada, Spain,GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Ignacio J. Molina
- Institute of Biopathology and Regenerative Medicine, Centre for Biomedical Research, University of Granada, Granada, Spain
| | - María José Pineda de las Infantas y Villatoro
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Granada, Spain,*Correspondence: Julio Alonso-Padilla, ; María José Pineda de las Infantas y Villatoro,
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain,*Correspondence: Julio Alonso-Padilla, ; María José Pineda de las Infantas y Villatoro,
| |
Collapse
|
10
|
Adamantane-Substituted Purine Nucleosides: Synthesis, Host-Guest Complexes with β-Cyclodextrin and Biological Activity. Int J Mol Sci 2022; 23:ijms232315143. [PMID: 36499470 PMCID: PMC9739181 DOI: 10.3390/ijms232315143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Purine nucleosides represent an interesting group of nitrogen heterocycles, showing a wide range of biological effects. In this study, we designed and synthesized a series of 6,9-disubstituted and 2,6,9-trisubstituted purine ribonucleosides via consecutive nucleophilic aromatic substitution, glycosylation, and deprotection of the ribofuranose unit. We prepared eight new purine nucleosides bearing unique adamantylated aromatic amines at position 6. Additionally, the ability of the synthesized purine nucleosides to form stable host-guest complexes with β-cyclodextrin (β-CD) was confirmed using nuclear magnetic resonance (NMR) and mass spectrometry (ESI-MS) experiments. The in vitro antiproliferative activity of purine nucleosides and their equimolar mixtures with β-CD was tested against two types of human tumor cell line. Six adamantane-based purine nucleosides showed an antiproliferative activity in the micromolar range. Moreover, their effect was only slightly suppressed by the presence of β-CD, which was probably due to the competitive binding of the corresponding purine nucleoside inside the β-CD cavity.
Collapse
|
11
|
Campagnaro GD. Purine Transporters as Efficient Carriers for Anti-kinetoplastid Molecules: 3'-Deoxytubercidin versus Trypanosomes. ACS Infect Dis 2022; 8:1727-1730. [PMID: 35925865 DOI: 10.1021/acsinfecdis.2c00356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
After a growing interest in the function of purine transporters in protozoa during the 1990s and early 2000s, the area experienced a lull phase. Recently, however, the potential of tubercidin derivatives, particularly 3'-deoxytubercidin, to cure Trypanosoma brucei infection seems to have started a new wave of interest in the subject, with a large number of newly designed compounds and extensive in vitro testing against T. brucei, Trypanosoma cruzi, and Leishmania spp. Understanding the biochemical properties of purine transporters and using them as drug carriers seem to be emerging once again as a valuable tactic in the fight against neglected diseases.
Collapse
Affiliation(s)
- Gustavo Daniel Campagnaro
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900 São Paulo, Brazil
| |
Collapse
|
12
|
Exploration of 6-methyl-7-(Hetero)Aryl-7-Deazapurine ribonucleosides as antileishmanial agents. Eur J Med Chem 2022; 237:114367. [DOI: 10.1016/j.ejmech.2022.114367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 11/18/2022]
|
13
|
Silva DG, Feijens P, Hendrickx R, Matheeussen A, Grey L, Caljon G, Maes L, Emery FS, Junker A. Development of Novel Isoindolone-Based Compounds against Trypanosoma brucei rhodesiense. ChemistryOpen 2021; 10:922-927. [PMID: 34553828 PMCID: PMC8459400 DOI: 10.1002/open.202100180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/02/2021] [Indexed: 12/03/2022] Open
Abstract
This study identified the isoindolone ring as a scaffold for novel agents against Trypanosoma brucei rhodesiense and explored the structure-activity relationships of various aromatic ring substitutions. The compounds were evaluated in an integrated in vitro screen. Eight compounds exhibited selective activity against T. b. rhodesiense (IC50 <2.2 μm) with no detectable side activity against T. cruzi and Leishmania infantum. Compound 20 showed low nanomolar potency against T. b. rhodesiense (IC50 =40 nm) and no toxicity against MRC-5 and PMM cell lines and may be regarded as a new lead template for agents against T. b. rhodesiense. The isoindolone-based compounds have the potential to progress into lead optimization in view of their highly selective in vitro potency, absence of cytotoxicity and acceptable metabolic stability. However, the solubility of the compounds represents a limiting factor that should be addressed to improve the physicochemical properties that are required to proceed further in the development of in vivo-active derivatives.
Collapse
Affiliation(s)
- Daniel G. Silva
- QHeteM - Laboratório de Química Heterocíclica e MedicinalSchool of Pharmaceutical Sciences of Ribeirão PretoUniversity of São PauloRibeirão PretoSão Paulo14040-903Brazil
- European Institute for Molecular Imaging (EIMI)Westphalian Wilhelms-University Münster48149MünsterGermany
| | - Pim‐Bart Feijens
- Laboratory of MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpUniversiteitsplein 1Wilrijk B2610Belgium
| | - Rik Hendrickx
- Laboratory of MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpUniversiteitsplein 1Wilrijk B2610Belgium
| | - An Matheeussen
- Laboratory of MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpUniversiteitsplein 1Wilrijk B2610Belgium
| | - Lucie Grey
- Institute for Pharmaceutical and Medicinal ChemistryWestphalian Wilhelms-University Münster48149MünsterGermany
| | - Guy Caljon
- Laboratory of MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpUniversiteitsplein 1Wilrijk B2610Belgium
| | - Louis Maes
- Laboratory of MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpUniversiteitsplein 1Wilrijk B2610Belgium
| | - Flavio S. Emery
- QHeteM - Laboratório de Química Heterocíclica e MedicinalSchool of Pharmaceutical Sciences of Ribeirão PretoUniversity of São PauloRibeirão PretoSão Paulo14040-903Brazil
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI)Westphalian Wilhelms-University Münster48149MünsterGermany
| |
Collapse
|
14
|
Acyclic nucleoside phosphonates with adenine nucleobase inhibit Trypanosoma brucei adenine phosphoribosyltransferase in vitro. Sci Rep 2021; 11:13317. [PMID: 34172767 PMCID: PMC8233378 DOI: 10.1038/s41598-021-91747-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/28/2021] [Indexed: 02/08/2023] Open
Abstract
All medically important unicellular protozoans cannot synthesize purines de novo and they entirely rely on the purine salvage pathway (PSP) for their nucleotide generation. Therefore, purine derivatives have been considered as a promising source of anti-parasitic compounds since they can act as inhibitors of the PSP enzymes or as toxic products upon their activation inside of the cell. Here, we characterized a Trypanosoma brucei enzyme involved in the salvage of adenine, the adenine phosphoribosyl transferase (APRT). We showed that its two isoforms (APRT1 and APRT2) localize partly in the cytosol and partly in the glycosomes of the bloodstream form (BSF) of the parasite. RNAi silencing of both APRT enzymes showed no major effect on the growth of BSF parasites unless grown in artificial medium with adenine as sole purine source. To add into the portfolio of inhibitors for various PSP enzymes, we designed three types of acyclic nucleotide analogs as potential APRT inhibitors. Out of fifteen inhibitors, four compounds inhibited the activity of the recombinant APRT1 with Ki in single µM values. The ANP phosphoramidate membrane-permeable prodrugs showed pronounced anti-trypanosomal activity in a cell-based assay, despite the fact that APRT enzymes are dispensable for T. brucei growth in vitro. While this suggests that the tested ANP prodrugs exert their toxicity by other means in T. brucei, the newly designed inhibitors can be further improved and explored to identify their actual target(s).
Collapse
|
15
|
Nguyen VH, Tichý M, Rožánková S, Pohl R, Downey AM, Doleželová E, Tloušťová E, Slapničková M, Zíková A, Hocek M. Synthesis and anti-trypanosomal activity of 3'-fluororibonucleosides derived from 7-deazapurine nucleosides. Bioorg Med Chem Lett 2021; 40:127957. [PMID: 33741462 DOI: 10.1016/j.bmcl.2021.127957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 10/21/2022]
Abstract
Trypanosoma brucei parasites cause Human African Trypanosomiasis and the current drugs for its treatment are often inefficient and toxic. This urges the need to development of new antitrypanosomal agents. We report the synthesis and biological profiling of 3'-deoxy-3'-fluororibonucleosides derived from 7-deazaadenine nucleosides bearing diverse substituents at position 7. They were synthesized through glycosylation of 6-chloro-7-bromo- or -7-iodo-7-deazapurine with protected 3'-fluororibose followed by cross-coupling reactions at position 7 and/or deprotection. Most of the title nucleosides displayed micromolar or submicromolar activity against Trypanosoma brucei brucei. The most active were the 7-bromo- and 7-iododerivatives which exerted double-digit nanomolar activity against T. b. brucei and T. b. gambiense and no cytotoxicity and thus represent promising candidates for further development.
Collapse
Affiliation(s)
- Van Hai Nguyen
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic; Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 8, CZ-12843 Prague 2, Czech Republic
| | - Michal Tichý
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic
| | - Samanta Rožánková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic
| | - Radek Pohl
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic
| | - A Michael Downey
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic
| | - Eva Doleželová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, CZ-37005 České Budějovice, Czech Republic
| | - Eva Tloušťová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic
| | - Martina Slapničková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, CZ-37005 České Budějovice, Czech Republic
| | - Alena Zíková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, CZ-37005 České Budějovice, Czech Republic.
| | - Michal Hocek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Czech Republic; Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 8, CZ-12843 Prague 2, Czech Republic.
| |
Collapse
|
16
|
Bouton J, Ferreira de Almeida Fiuza L, Cardoso Santos C, Mazzarella MA, Soeiro MDNC, Maes L, Karalic I, Caljon G, Van Calenbergh S. Revisiting Pyrazolo[3,4- d]pyrimidine Nucleosides as Anti- Trypanosoma cruzi and Antileishmanial Agents. J Med Chem 2021; 64:4206-4238. [PMID: 33784107 DOI: 10.1021/acs.jmedchem.1c00135] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Chagas disease and visceral leishmaniasis are two neglected tropical diseases responsible for numerous deaths around the world. For both, current treatments are largely inadequate, resulting in a continued need for new drug discovery. As both kinetoplastid parasites are incapable of de novo purine synthesis, they depend on purine salvage pathways that allow them to acquire and process purines from the host to meet their demands. Purine nucleoside analogues therefore constitute a logical source of potential antiparasitic agents. Earlier optimization efforts of the natural product tubercidin (7-deazaadenosine) involving modifications to the nucleobase 7-position and the ribofuranose 3'-position led to analogues with potent anti-Trypanosoma brucei and anti-Trypanosoma cruzi activities. In this work, we report the design and synthesis of pyrazolo[3,4-d]pyrimidine nucleosides with 3'- and 7-modifications and assess their potential as anti-Trypanosoma cruzi and antileishmanial agents. One compound was selected for in vivo evaluation in an acute Chagas disease mouse model.
Collapse
Affiliation(s)
- Jakob Bouton
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Ludmila Ferreira de Almeida Fiuza
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ), Fundação Oswaldo Cruz, Rio de Janeiro, Avenida Brasil 4365, Manguinhos, 21040-360 Rio de Janeiro, Brazil
| | - Camila Cardoso Santos
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ), Fundação Oswaldo Cruz, Rio de Janeiro, Avenida Brasil 4365, Manguinhos, 21040-360 Rio de Janeiro, Brazil
| | - Maria Angela Mazzarella
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, Perugia 06100, Italy
| | - Maria de Nazaré Correia Soeiro
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ), Fundação Oswaldo Cruz, Rio de Janeiro, Avenida Brasil 4365, Manguinhos, 21040-360 Rio de Janeiro, Brazil
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Izet Karalic
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| |
Collapse
|