1
|
Majoor A, Michel G, Marty P, Boyer L, Pomares C. Leishmaniases: Strategies in treatment development. Parasite 2025; 32:18. [PMID: 40043198 PMCID: PMC11882135 DOI: 10.1051/parasite/2025009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Leishmaniases are vector-borne parasitic diseases that pose a threat to over 1 billion people worldwide. The parasites target cells of the reticulohistiocytic system, such as macrophages, where they replicate. The disease manifests in various forms, ranging from localized cutaneous leishmaniasis to life-threatening visceral forms, which are fatal in 95% of cases without treatment. Current treatments rely on the invasive administration of toxic and expensive drugs that are increasingly encountering resistance. Therefore, finding alternative treatments for this disease is imperative. This literature review focuses on recent advancements in alternative treatments and aims to present the various strategies designed to address current limitations, including cost, toxicity, off-target effects, administration routes, and the emergence of drug resistance. Starting with an overview of the existing approved treatments and their specific limitations, we categorize treatment development strategies into five key sections: (i) combination therapies using existing approved treatments to enhance efficacy and reduce resistance; (ii) nanoparticle formulations, which enable targeted delivery to infected organs and improved therapeutic efficiency; (iii) drug repositioning, a strategy that has already contributed to the approval of over half of current therapeutic compounds; (iv) immunomodulation, used in conjunction with standard chemotherapies to enhance treatment efficacy and lower relapse rates; and (v) ethnobotanicals, which have demonstrated promising in vitro results by combining low toxicity, immunomodulatory properties, and potent anti-parasitic effects. In summary, this review outlines current strategies in treatment development, emphasizing their advantages over conventional therapies while acknowledging their limitations.
Collapse
Affiliation(s)
- Alissa Majoor
- Université Côte d’Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) BP 23194 06204 Nice Cedex 3 France
| | - Grégory Michel
- Université Côte d’Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) BP 23194 06204 Nice Cedex 3 France
| | - Pierre Marty
- Université Côte d’Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) BP 23194 06204 Nice Cedex 3 France
- Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet CS 23079 06202 Nice Cedex 3 France
| | - Laurent Boyer
- Université Côte d’Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) BP 23194 06204 Nice Cedex 3 France
| | - Christelle Pomares
- Université Côte d’Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) BP 23194 06204 Nice Cedex 3 France
- Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet CS 23079 06202 Nice Cedex 3 France
| |
Collapse
|
2
|
Badenhorst E, Aucamp J, Kannigadu C, Janse van Rensburg HD, Suganuma K, N’Da DD. Synthesis and in vitro antitrypanosomatid activity of novel 5-nitroindole-rhodanine conjugates. Future Med Chem 2025; 17:557-573. [PMID: 39995114 PMCID: PMC11901381 DOI: 10.1080/17568919.2025.2470110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
AIM Trypanosomatid diseases, leishmaniasis and trypanosomiasis are vector-borne parasitic diseases that can cause death and catastrophic economic losses for millions of people. The growing resistance of trypanosomatid parasites to current treatments highlights the urgent need for new therapeutic agents. This study explored 5-nitroindole-rhodanine conjugates to identify promising new compounds with the potential for future development as antitrypanosomatid treatments. MATERIALS AND METHODS The conjugates were synthesized in a multi-step process and evaluated in vitro for antileishmanial activity against Leishmania (L.) donovani and L. major strains. Cytotoxicity was assessed on Vero and THP-1 cells. Due to the taxonomic relation to Trypanosoma spp. the compounds were also screened for in vitro activity against species that cause zoonotic trypanosomiasis. RESULTS AND CONCLUSION Several hits were found with leishmanicidal activity against both L. donovani and L. major strains. Of these, 3d was identified as a potential early lead that exhibited nanomolar cidal activity against L. major, and greater selectivity than the reference drug amphotericin B. However, the compounds did not have similar activity levels against Trypanosoma spp. Hence, these compounds should be further investigated for their mechanism of action and in vivo antileishmanial activity to determine their potential as a leishmaniasis treatment.
Collapse
Affiliation(s)
- Emce Badenhorst
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Christina Kannigadu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | | | - Keisuke Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - David D. N’Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
3
|
Sundar S, Madhukar P, Kumar R. Anti-leishmanial therapies: overcoming current challenges with emerging therapies. Expert Rev Anti Infect Ther 2025; 23:159-180. [PMID: 39644325 DOI: 10.1080/14787210.2024.2438627] [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: 07/22/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Leishmaniasis, including visceral, cutaneous, and mucocutaneous forms, present a major health challenge in tropical regions. Current antileishmanial medications has significant limitations, creating a critical need for novel therapies that are safe and cost-effective with a shorter duration of treatment. AREAS COVERED This review explores the critical aspects of existing antileishmanial therapy and targets for future therapeutic developments. It emphasizes the need for new treatment options due to drug resistance, low success rates, toxicity, and high prices associated with current medications. The different forms of leishmaniasis, their clinical manifestations, the challenges associated with their treatment and emerging treatment options are explored in detail. EXPERT OPINION The first anti-leishmanial drug pentavalent antimony (SbV) was invented more than 100 years back. Since then, this compound has been used for all forms of leishmaniasis worldwide. For more than 70-80 years after discovery of SbV, no new antileishmanial drugs were developed, reflecting the lack of interest from academia or pharma industry. All three new treatments (Amphotericin-B, paromomycin and miltefosine) which underwent the clinical trials were repurposed drugs. The current pipeline for antileishmanial drugs is empty, with LXE 408 being the only potential drug reaching phase II clinical trial.
Collapse
Affiliation(s)
- Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Prasoon Madhukar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
4
|
Sundar S, Singh VK, Agrawal N, Singh OP, Kumar R. Investigational new drugs for the treatment of leishmaniasis. Expert Opin Investig Drugs 2024; 33:1029-1046. [PMID: 39225742 DOI: 10.1080/13543784.2024.2400139] [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: 12/29/2023] [Revised: 08/05/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Over the past 20 years, significant progress has been made in anti-leishmanial therapy. Three new drugs/formulations are available for the treatment of various forms of leishmaniasis, namely oral miltefosine, paromomycin and liposomal amphotericin B. However, these advances in drug development have added considerable complexity for clinicians including toxicity, emergence of resistance and decreased sensitivity of available drugs. The development of newer drugs with less toxicity and more efficacy is urgently needed. AREAS COVERED This review comprehensively examines the latest developments and current status of antileishmanial drugs for the treatment of leishmaniasis across the world. Several new investigational drugs that showed anti-leishmanial activity under in vitro or in vivo conditions and either underwent the phase-I/II clinical trials or are on the verge of entering the trials were reviewed. We also delve into the challenges of drug resistance and discuss the emergence of new and effective antileishmanial compounds. EXPERT OPINION The available treatments for leishmaniasis are limited in number, toxic, expensive, and demand extensive healthcare resources. Every available antileishmanial drug is associated with several disadvantages, such as drug resistance and toxicity or high cost. Miltefosine is potentially teratogenic. New antileishmanial drugs/treatment modalities are sorely needed for expanding future treatment options.
Collapse
Affiliation(s)
- Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Neha Agrawal
- Department of Medicine, University of Florida, Jacksonville, FL, USA
| | - Om Prakash Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
5
|
Santamaría-Aguirre J, Jacho D, Méndez MA, Poveda A, Carrión J, Fanarraga ML. Solid Lipid Nanoparticles Enhancing the Leishmanicidal Activity of Delamanid. Pharmaceutics 2023; 16:41. [PMID: 38258053 PMCID: PMC10818933 DOI: 10.3390/pharmaceutics16010041] [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: 11/06/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
Leishmaniasis, a zoonotic parasitic disease transmitted by infected sandflies, impacts nearly 1 million people yearly and is endemic in many countries across Asia, Africa, the Americas, and the Mediterranean; despite this, it remains a neglected disease with limited effective treatments, particularly in impoverished communities with limited access to healthcare. This study aims to repurpose approved drugs for an affordable leishmaniasis treatment. After the screening of potential drug candidates by reviewing databases and utilizing molecular docking analysis, delamanid was chosen to be incorporated into solid lipid nanoparticles (SLNPs). Both in cellulo and in vivo tests confirmed the successful payload release within macrophages and through the epidermis following topical application on murine skin. The evaluation of macrophages infected with L. infantum amastigotes showed that the encapsulated delamanid exhibited greater leishmanicidal activity compared with the free drug. The process of encapsulating delamanid in SLNPs, as demonstrated in this study, places a strong emphasis on employing minimal technology, ensuring energy efficiency, cost-effectiveness, and reproducibility. It enables consistent, low-cost production of nanomedicines, even on a small scale, offering a promising step toward more accessible and effective leishmaniasis treatments.
Collapse
Affiliation(s)
- Javier Santamaría-Aguirre
- Departamento de Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain
- Grupo de Nanomedicina, Instituto Valdecilla—IDIVAL, 39011 Santander, Spain
- Grupo de Investigación en Biodiversidad, Zoonosis y Salud Pública (GIBCIZ), Instituto de Investigación en Zoonosis (CIZ), Facultad de Ciencias Químicas (FCQ), Universidad Central del Ecuador, Quito 170521, Ecuador
| | - Daniela Jacho
- Facultad de Ciencias Químicas (FCQ), Universidad Central del Ecuador, Quito 170521, Ecuador
| | - Miguel A. Méndez
- Departamento de Ingeniería Química, Instituto de Simulación Computacional (ISC-USFQ), Universidad San Francisco de Quito USFQ, Quito 170157, Ecuador
| | - Ana Poveda
- Grupo de Investigación en Biodiversidad, Zoonosis y Salud Pública (GIBCIZ), Instituto de Investigación en Zoonosis (CIZ), Facultad de Ciencias Químicas (FCQ), Universidad Central del Ecuador, Quito 170521, Ecuador
| | - Javier Carrión
- Grupo ICPVet, Departamento Sanidad Animal, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Mónica L. Fanarraga
- Departamento de Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain
- Grupo de Nanomedicina, Instituto Valdecilla—IDIVAL, 39011 Santander, Spain
| |
Collapse
|
6
|
Abdullah S, Ganguly S. An overview of imidazole and its analogues as potent anticancer agents. Future Med Chem 2023; 15:1621-1646. [PMID: 37727960 DOI: 10.4155/fmc-2023-0020] [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: 09/21/2023] Open
Abstract
The quest for novel, physiologically active imidazoles remains an exciting topic of research among medicinal chemists. The imidazole ring is a five-membered aromatic heterocycle that is found in both natural and synthesized compounds. Multiple anticancer drug classes are currently available on the market, but concerns including toxicity, limited efficacy and solubility have lowered the overall therapeutic index. Therefore, the hunt for new potential chemotherapeutic agents persists. The development of imidazole as a reliable and safer alternative to anticancer treatment is generating much attention among experts. Tubulin or microtubule polymerization inhibition and changes in the structure and function of DNA, VEGF, topoisomerase, kinases, histone deacetylases and certain other proteins that affect gene expression are among the putative targets.
Collapse
Affiliation(s)
- Salik Abdullah
- Department of Pharmaceutical Sciences, Birla Institute of Technology, Mesra, Jharkhand, 835215, India
| | - Swastika Ganguly
- Department of Pharmaceutical Sciences, Birla Institute of Technology, Mesra, Jharkhand, 835215, India
| |
Collapse
|
7
|
Ernest JP, Goh JJN, Strydom N, Wang Q, van Wijk RC, Zhang N, Deitchman A, Nuermberger E, Savic RM. Translational predictions of phase 2a first-in-patient efficacy studies for antituberculosis drugs. Eur Respir J 2023; 62:2300165. [PMID: 37321622 PMCID: PMC10469274 DOI: 10.1183/13993003.00165-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Phase 2a trials in tuberculosis typically use early bactericidal activity (EBA), the decline in sputum CFU over 14 days, as the primary end-point for testing the efficacy of drugs as monotherapy. However, the cost of phase 2a trials can range from USD 7 million to USD 19.6 million on average, while >30% of drugs fail to progress to phase 3. Better utilising pre-clinical data to predict and prioritise the most likely drugs to succeed will thus help to accelerate drug development and reduce costs. We aim to predict clinical EBA using pre-clinical in vivo pharmacokinetic (PK)-pharmacodynamic (PD) data and a model-based translational pharmacology approach. METHODS AND FINDINGS First, mouse PK, PD and clinical PK models were compiled. Second, mouse PK-PD models were built to derive an exposure-response relationship. Third, translational prediction of clinical EBA studies was performed using mouse PK-PD relationships and informed by clinical PK models and species-specific protein binding. Presence or absence of clinical efficacy was accurately predicted from the mouse model. Predicted daily decreases of CFU in the first 2 days of treatment and between day 2 and day 14 were consistent with clinical observations. CONCLUSION This platform provides an innovative solution to inform or even replace phase 2a EBA trials, to bridge the gap between mouse efficacy studies and phase 2b and phase 3 trials, and to substantially accelerate drug development.
Collapse
Affiliation(s)
- Jacqueline P Ernest
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Shared authorship ordered alphabetically
| | - Janice Jia Ni Goh
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Shared authorship ordered alphabetically
| | - Natasha Strydom
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Shared authorship ordered alphabetically
| | - Qianwen Wang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Shared authorship ordered alphabetically
| | - Rob C van Wijk
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Shared authorship ordered alphabetically
| | - Nan Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Shared authorship ordered alphabetically
| | - Amelia Deitchman
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Eric Nuermberger
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MA, USA
| | - Rada M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
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
|
9
|
Lanni F, Wijnant GJ, Xie M, Osiecki P, Dartois V, Sarathy JP. Adaptation to the intracellular environment of primary human macrophages influences drug susceptibility of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2023; 139:102318. [PMID: 36889104 DOI: 10.1016/j.tube.2023.102318] [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: 08/29/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 01/24/2023]
Abstract
As a facultative intracellular pathogen, M. tuberculosis (Mtb) is highly adapted to evading antibacterial mechanisms in phagocytic cells. Both the macrophage and pathogen experience transcriptional and metabolic changes from the onset of phagocytosis. To account for this interaction in the assessment of intracellular drug susceptibility, we allowed a 3-day preadaptation phase post-macrophage infection prior to drug treatment. We found that intracellular Mtb in human monocyte-derived macrophages (MDM) presents dramatic alterations in susceptibility to isoniazid, sutezolid, rifampicin and rifapentine when compared to axenic culture. Infected MDM gradually accumulate lipid bodies, adopting a characteristic appearance reminiscent of foamy macrophages in granulomas. Furthermore, TB granulomas in vivo develop hypoxic cores with decreasing oxygen tension gradients across their radii. Accordingly, we evaluated the effects of hypoxia on preadapted intracellular Mtb in our MDM model. We observed that hypoxia induced greater lipid body formation and no additional shifts in drug tolerance, suggesting that the adaptation of intracellular Mtb to baseline host cell conditions under normoxia dominates changes to intracellular drug susceptibility. Using unbound plasma concentrations in patients as surrogates for free drug concentrations in lung interstitial fluid, we estimate that intramacrophage Mtb in granulomas are exposed to bacteriostatic concentrations of most study drugs.
Collapse
Affiliation(s)
- Faye Lanni
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Gert-Jan Wijnant
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Min Xie
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Paulina Osiecki
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States; Hackensack School of Medicine, Department of Medical Sciences, 123, Metro Boulevard, Nutley, NJ, 07110, United States
| | - Jansy P Sarathy
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States.
| |
Collapse
|
10
|
Urán Landaburu L, Didier Garnham M, Agüero F. Targeting trypanosomes: how chemogenomics and artificial intelligence can guide drug discovery. Biochem Soc Trans 2023; 51:195-206. [PMID: 36606702 DOI: 10.1042/bst20220618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
Trypanosomatids are protozoan parasites that cause human and animal neglected diseases. Despite global efforts, effective treatments are still much needed. Phenotypic screens have provided several chemical leads for drug discovery, but the mechanism of action for many of these chemicals is currently unknown. Recently, chemogenomic screens assessing the susceptibility or resistance of parasites carrying genome-wide modifications started to define the mechanism of action of drugs at large scale. In this review, we discuss how genomics is being used for drug discovery in trypanosomatids, how integration of chemical and genomics data from these and other organisms has guided prioritisations of candidate therapeutic targets and additional chemical starting points, and how these data can fuel the expansion of drug discovery pipelines into the era of artificial intelligence.
Collapse
Affiliation(s)
- Lionel Urán Landaburu
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Mercedes Didier Garnham
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| |
Collapse
|
11
|
Ernest JP, Ni Goh JJ, Strydom N, Wang Q, van Wijk RC, Zhang N, Deitchman A, Nuermberger E, Savic RM. Translational predictions of phase 2a first-in-patient efficacy studies for antituberculosis drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524608. [PMID: 36711493 PMCID: PMC9882354 DOI: 10.1101/2023.01.18.524608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Phase 2a trials in tuberculosis typically use early bactericidal activity (EBA), the decline in sputum colony forming units (CFU) over 14 days, as the primary outcome for testing the efficacy of drugs as monotherapy. However, the cost of phase 2a trials can range from 7 to 19.6 million dollars on average, while more than 30% of drugs fail to progress to phase 3. Better utilizing preclinical data to predict and prioritize the most likely drugs to succeed will thus help accelerate drug development and reduce costs. We aim to predict clinical EBA using preclinical in vivo pharmacokinetic-pharmacodynamic (PKPD) data and a model-based translational pharmacology approach. Methods and Findings First, mouse PK, PD and clinical PK models were compiled. Second, mouse PKPD models were built to derive an exposure response relationship. Third, translational prediction of clinical EBA studies was performed using mouse PKPD relationships and informed by clinical PK models and species-specific protein binding. Presence or absence of clinical efficacy was accurately predicted from the mouse model. Predicted daily decreases of CFU in the first 2 days of treatment and between day 2 and day 14 were consistent with clinical observations. Conclusion This platform provides an innovative solution to inform or even replace phase 2a EBA trials, to bridge the gap between mouse efficacy studies and phase 2b and phase 3 trials, and to substantially accelerate drug development.
Collapse
|
12
|
Gopu B, Kour P, Pandian R, Singh K. Insights into the drug screening approaches in leishmaniasis. Int Immunopharmacol 2023; 114:109591. [PMID: 36700771 DOI: 10.1016/j.intimp.2022.109591] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/25/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Leishmaniasis, a tropically neglected disease, is responsible for the high mortality and morbidity ratio in poverty-stricken areas. Currently, no vaccine is available for the complete cure of the disease. Current chemotherapeutic regimens face the limitations of drug resistance and toxicity concerns indicating a great need to develop better chemotherapeutic leads that are orally administrable, potent, non-toxic, and cost-effective. The anti-leishmanial drug discovery process accelerated the desire for large-scale drug screening assays and high-throughput screening (HTS) technology to identify new chemo-types that can be used as potential drug molecules to control infection. Using the HTS approach, about one million compounds can be screened daily within the shortest possible time for biological activity using automation tools, miniaturized assay formats, and large-scale data analysis. Classical and modern in vitro screening assays have led to the progression of active compounds further to ex vivo and in vivo studies. In the present review, we emphasized on the HTS approaches employed in the leishmanial drug discovery program. Recent in vitro screening assays are widely explored to discover new chemical scaffolds. Developing appropriate experimental animal models and their related techniques is necessary to understand the pathophysiological processes and disease host responses, paving the way for unraveling novel therapies against leishmaniasis.
Collapse
Affiliation(s)
- Boobalan Gopu
- Animal House Facility, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Parampreet Kour
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Ramajayan Pandian
- Animal House Facility, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
13
|
Ang CW, Lee BM, Jackson CJ, Wang Y, Franzblau SG, Francisco AF, Kelly JM, Bernhardt PV, Tan L, West NP, Sykes ML, Hinton AO, Bolisetti R, Avery VM, Cooper MA, Blaskovich MA. Nitroimidazopyrazinones with Oral Activity against Tuberculosis and Chagas Disease in Mouse Models of Infection. J Med Chem 2022; 65:13125-13142. [DOI: 10.1021/acs.jmedchem.2c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chee Wei Ang
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Science, Monash University Malaysia, Subang Jaya, 47500 Selangor, Malaysia
| | - Brendon M. Lee
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, United States
| | - Colin J. Jackson
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Paul V. Bernhardt
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Melissa L. Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Alexandra O. Hinton
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Raghu Bolisetti
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Matthew A. Cooper
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mark A.T. Blaskovich
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
14
|
Jain S, Sahu U, Kumar A, Khare P. Metabolic Pathways of Leishmania Parasite: Source of Pertinent Drug Targets and Potent Drug Candidates. Pharmaceutics 2022; 14:pharmaceutics14081590. [PMID: 36015216 PMCID: PMC9416627 DOI: 10.3390/pharmaceutics14081590] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a tropical disease caused by a protozoan parasite Leishmania that is transmitted via infected female sandflies. At present, leishmaniasis treatment mainly counts on chemotherapy. The currently available drugs against leishmaniasis are costly, toxic, with multiple side effects, and limitations in the administration route. The rapid emergence of drug resistance has severely reduced the potency of anti-leishmanial drugs. As a result, there is a pressing need for the development of novel anti-leishmanial drugs with high potency, low cost, acceptable toxicity, and good pharmacokinetics features. Due to the availability of preclinical data, drug repurposing is a valuable approach for speeding up the development of effective anti-leishmanial through pointing to new drug targets in less time, having low costs and risk. Metabolic pathways of this parasite play a crucial role in the growth and proliferation of Leishmania species during the various stages of their life cycle. Based on available genomics/proteomics information, known pathways-based (sterol biosynthetic pathway, purine salvage pathway, glycolysis, GPI biosynthesis, hypusine, polyamine biosynthesis) Leishmania-specific proteins could be targeted with known drugs that were used in other diseases, resulting in finding new promising anti-leishmanial therapeutics. The present review discusses various metabolic pathways of the Leishmania parasite and some drug candidates targeting these pathways effectively that could be potent drugs against leishmaniasis in the future.
Collapse
Affiliation(s)
- Surbhi Jain
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal 462026, Madhya Pradesh, India; (S.J.); (U.S.)
| | - Utkarsha Sahu
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal 462026, Madhya Pradesh, India; (S.J.); (U.S.)
- Division of Synthetic Biology, Absolute Foods, Plot 68, Sector 44, Gurugram 122003, Haryana, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur 492010, Chhattisgarh, India
- Correspondence: or (A.K.); (P.K.)
| | - Prashant Khare
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal 462026, Madhya Pradesh, India; (S.J.); (U.S.)
- Division of Synthetic Biology, Absolute Foods, Plot 68, Sector 44, Gurugram 122003, Haryana, India
- Correspondence: or (A.K.); (P.K.)
| |
Collapse
|
15
|
Functionalized Nitroimidazole Scaffold Construction and Their Pharmaceutical Applications: A 1950–2021 Comprehensive Overview. Pharmaceuticals (Basel) 2022; 15:ph15050561. [PMID: 35631389 PMCID: PMC9144801 DOI: 10.3390/ph15050561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Nitroimidazole represents one of the most essential and unique scaffolds in drug discovery since its discovery in the 1950s. It was K. Maeda in Japan who reported in 1953 the first nitroimidazole as a natural product from Nocardia mesenterica with antibacterial activity, which was later identified as Azomycin 1 (2-nitroimidazole) and remained in focus until now. This natural antibiotic was the starting point for synthesizing numerous analogs and regio-isomers, leading to several life-saving drugs and clinical candidates against a number of diseases, including infections (bacterial, viral, parasitic) and cancers, as well as imaging agents in medicine/diagnosis. In the present decade, the nitroimidazole scaffold has again been given two life-saving drugs (Delamanid and Pretomanid) used to treat MDR (multi-drug resistant) tuberculosis. Keeping in view the highly successful track-record of the nitroimidazole scaffold in providing breakthrough therapeutic drugs, this comprehensive review focuses explicitly on presenting the activity profile and synthetic chemistry of functionalized nitroimidazole (2-, 4- and 5-nitroimidazoles as well as the fused nitroimidazoles) based drugs and leads published from 1950 to 2021. The present review also presents the miscellaneous examples in each class. In addition, the mutagenic profile of nitroimidazole-based drugs and leads and derivatives is also discussed.
Collapse
|
16
|
Liu J, Lin W, Sorochinsky AE, Butler G, Landa A, Han J, Soloshonok VA. Successful trifluoromethoxy-containing pharmaceuticals and agrochemicals. J Fluor Chem 2022. [DOI: 10.1016/j.jfluchem.2022.109978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Ghanbariasad A, Emami L, Zarenezhad E, Behrouz S, Zarenezhad A, Soltani Rad MN. Synthesis, Biological Evaluation and In silico Studies of 1, 2, 3-triazolyl- Metronidazole Derivatives Against Leishmania Major. NEW J CHEM 2022. [DOI: 10.1039/d2nj00226d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The simple and effective approach for the preparation, of 1,2,3-triazolyl-based metronidazole hybrid analogues as promising anti-leishmania agents using of [CuL-SiO-HA] as a catalyst were described. The catalyst was fully characterized...
Collapse
|
18
|
Khoshnood S, Taki E, Sadeghifard N, Kaviar VH, Haddadi MH, Farshadzadeh Z, Kouhsari E, Goudarzi M, Heidary M. Mechanism of Action, Resistance, Synergism, and Clinical Implications of Delamanid Against Multidrug-Resistant Mycobacterium tuberculosis. Front Microbiol 2021; 12:717045. [PMID: 34690963 PMCID: PMC8529252 DOI: 10.3389/fmicb.2021.717045] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/02/2021] [Indexed: 11/21/2022] Open
Abstract
Multidrug-resistant (MDR) isolates of Mycobacterium tuberculosis (MTB) remain a primary global threat to the end of tuberculosis (TB) era. Delamanid (DLM) is a nitro-dihydro-imidazooxazole derivative utilized to treat MDR-TB. DLM has distinct mechanism of action, inhibiting methoxy- and keto-mycolic acid (MA) synthesis through the F420 coenzyme mycobacteria system and generating nitrous oxide. While DLM resistance among MTB strains is uncommon, there are increasing reports in Asia and Europe, and such resistance will prolong the treatment courses of patients infected with MDR-TB. In this review, we address the antimycobacterial properties of DLM, report the global prevalence of DLM resistance, discuss the synergism of DLM with other anti-TB drugs, and evaluate the documented clinical trials to provide new insights into the clinical use of this antibiotic.
Collapse
Affiliation(s)
- Saeed Khoshnood
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Elahe Taki
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nourkhoda Sadeghifard
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Vahab Hassan Kaviar
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Zahra Farshadzadeh
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ebrahim Kouhsari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
19
|
Pacheco JDS, Costa DDS, Cunha-Júnior EF, Andrade-Neto VV, Fairlamb AH, Wyllie S, Goulart MOF, Santos DC, Silva TL, Alves MA, Costa PRR, Dias AG, Torres-Santos EC. Monocyclic Nitro-heteroaryl Nitrones with Dual Mechanism of Activation: Synthesis and Antileishmanial Activity. ACS Med Chem Lett 2021; 12:1405-1412. [PMID: 34531949 DOI: 10.1021/acsmedchemlett.1c00193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/05/2021] [Indexed: 12/17/2022] Open
Abstract
5-Nitro-furan nitrones (1) and 5-nitro-thiophene nitrones (2) were synthesized in one step. Compounds 1a-c had the most potent leishmanicidal activity against intracellular amastigote forms of Leishmania amazonensis and L. infantum (from 0.019 to 2.76 μM), with excellent selectivity (from 39 to 5673). The comparison of the leishmanicidal activity in promastigotes of wild type L. donovani with those overexpressing nitroreductases NRT1 or NRT2 shows that 1a,b are activated by both, which could slow the development of resistance. Their redox potential (E redox) obtained by cyclic voltammetry (-0.67 and -0.62 V) shows that the reduction of the nitro group is modulated by the nitrone group. Oral administration of 1b to mice infected by L. infantum reduced the parasite load on the spleen by 76.6 and 95.0% with doses of 50 and 100 mg/kg, respectively, administered twice a day, for 5 days. In the liver, the parasite load suppression was above 75% with either treatment.
Collapse
Affiliation(s)
- Juliana da Silva Pacheco
- FIOCRUZ, Instituto Oswaldo Cruz, Laboratório de Bioquímica de Tripanosomatídeos, Rio de Janeiro, RJ, Brazil
| | - Débora de Souza Costa
- Universidade Federal do Rio de Janeiro, Instituto de Pesquisas de Produtos Naturais, Laboratório de Química Bioorgânica, Rio de Janeiro, RJ, Brazil
| | | | - Valter Viana Andrade-Neto
- FIOCRUZ, Instituto Oswaldo Cruz, Laboratório de Bioquímica de Tripanosomatídeos, Rio de Janeiro, RJ, Brazil
| | - Alan H. Fairlamb
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, Scotland, United Kingdom
| | - Susan Wyllie
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, Scotland, United Kingdom
| | - Marília O. F. Goulart
- Universidade Federal de Alagoas, Instituto de Química e Biotecnologia, Maceió, AL, Brazil
| | - Danyelle C. Santos
- Universidade Federal de Alagoas, Instituto de Química e Biotecnologia, Maceió, AL, Brazil
| | - Thaissa L. Silva
- Universidade Federal de Alagoas, Núcleo de Ciências Exatas, Campus de Arapiraca, Arapiraca, AL, Brazil
| | - Marina A. Alves
- Universidade Federal do Rio de Janeiro, Laboratório de Apoio ao Desenvolvimento Tecnológico, Rio de Janeiro, RJ, Brazil
| | - Paulo R. R. Costa
- Universidade Federal do Rio de Janeiro, Instituto de Pesquisas de Produtos Naturais, Laboratório de Química Bioorgânica, Rio de Janeiro, RJ, Brazil
| | - Ayres G. Dias
- Universidade do Estado do Rio de Janeiro, Centro de Tecnologia e Ciências, Departamento de Química Orgânica, Rio de Janeiro, RJ, Brazil
| | - Eduardo Caio Torres-Santos
- FIOCRUZ, Instituto Oswaldo Cruz, Laboratório de Bioquímica de Tripanosomatídeos, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
20
|
Blanco Z, Mijares MR, Ramírez H, Fernandez-Moreira E, Oviedo HJ, Rodríguez NM, Charris JE. In vitro evaluation and in vivo efficacy of nitroimidazole-sulfanyl ethyl derivatives against Leishmania (V.) braziliensis and Leishmania (L.) mexicana. Parasitol Res 2021; 120:3307-3317. [PMID: 34370070 DOI: 10.1007/s00436-021-07266-w] [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: 04/12/2021] [Accepted: 07/21/2021] [Indexed: 11/24/2022]
Abstract
The aim of this study was to synthesize several small molecules of the type 5-nitroimidazole-sulfanyl and evaluate biological properties against the main Leishmania species that cause cutaneous leishmaniasis in Venezuela. Final compounds 4-7 were generated through simple nucleophilic substitution of 1-(2-chloroethyl)-2-methyl-5-nitroimidazole 3 with 2-mercaptoethanol, 1-methyl-2-mercaptoethanol, and 2-thyolacetic acid derivative. Compound 8 was synthesized via a coupling reaction between 7 and (S)-Methyl 2-amino-4-methylpentanoate hydrochloride. The inhibitory concentrations of (3, 4, 7, 8) against Leishmania (L.) mexicana and (V.) braziliensis in promastigotes and experimentally infected macrophages were determined by in vitro activity assays. Compounds 7 and 8 shown high activity against both species of Leishmania and were selected for the in vivo evaluation. Animals were infected with promastigotes of the two species and divided into four groups of ten (10) animals and a control group. Intralesional injection way was used for the treatment. The parasitological diagnostic after treatment was obtained by PCR using species specific oligonucleotides. The two Leishmania species were susceptible to compounds 7 and 8 in vivo assays. The results indicated that both compounds reduce significantly (96%) the size of the lesion and cure 63% of the mice infected with L (L) mexicana or L (V) braziliensis as was determined by PCR. The results are indicating that both compounds may represent an alternative treatment for these two Leishmania species.
Collapse
Affiliation(s)
- Zuleima Blanco
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, Los Chaguaramos 1041-A, Caracas, 47206, Venezuela
| | - Michael R Mijares
- Biotechnology Unit, Faculty of Pharmacy, Central University of Venezuela, Los Chaguaramos 1041-A, Caracas, 47206, Venezuela
| | - Hegira Ramírez
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, Los Chaguaramos 1041-A, Caracas, 47206, Venezuela.,Facultad de Medicina, Universidad de Las Américas, Quito, Ecuador
| | | | - Henry J Oviedo
- Institute of Biomedicine, Faculty of Medicine, Central University of Venezuela, 1010-A, 4043, Caracas, Venezuela
| | - Noris M Rodríguez
- Institute of Biomedicine, Faculty of Medicine, Central University of Venezuela, 1010-A, 4043, Caracas, Venezuela
| | - Jaime E Charris
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, Los Chaguaramos 1041-A, Caracas, 47206, Venezuela.
| |
Collapse
|
21
|
Rice AM, Long Y, King SB. Nitroaromatic Antibiotics as Nitrogen Oxide Sources. Biomolecules 2021; 11:267. [PMID: 33673069 PMCID: PMC7918234 DOI: 10.3390/biom11020267] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/21/2022] Open
Abstract
Nitroaromatic antibiotics show activity against anaerobic bacteria and parasites, finding use in the treatment of Heliobacter pylori infections, tuberculosis, trichomoniasis, human African trypanosomiasis, Chagas disease and leishmaniasis. Despite this activity and a clear need for the development of new treatments for these conditions, the associated toxicity and lack of clear mechanisms of action have limited their therapeutic development. Nitroaromatic antibiotics require reductive bioactivation for activity and this reductive metabolism can convert the nitro group to nitric oxide (NO) or a related reactive nitrogen species (RNS). As nitric oxide plays important roles in the defensive immune response to bacterial infection through both signaling and redox-mediated pathways, defining controlled NO generation pathways from these antibiotics would allow the design of new therapeutics. This review focuses on the release of nitrogen oxide species from various nitroaromatic antibiotics to portend the increased ability for these compounds to positively impact infectious disease treatment.
Collapse
Affiliation(s)
| | | | - S. Bruce King
- Department of Chemistry and Biochemistry, Wake Forest University, Winston-Salem, NC 27101, USA; (A.M.R.); (Y.L.)
| |
Collapse
|
22
|
Souza Silva JA, Tunes LG, Coimbra RS, Ascher DB, Pires DEV, Monte-Neto RL. Unveiling six potent and highly selective antileishmanial agents via the open source compound collection 'Pathogen Box' against antimony-sensitive and -resistant Leishmania braziliensis. Biomed Pharmacother 2020; 133:111049. [PMID: 33378956 DOI: 10.1016/j.biopha.2020.111049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
Despite all efforts to provide new chemical entities to tackle leishmaniases, we are still dependent on a the limited drug arsenal, together with drawbacks like toxicity and drug-resistant parasites. Collaborative drug discovery emerged as an option to speed up the way to find alternative antileishmanial agents. This is the case of Medicines for Malaria Ventures - MMV, that promotes an open source drug discovery initiative to fight diseases worldwide. Here, we screened 400 compounds from 'Pathogen Box' (PBox) collection against Leishmania braziliensis, the main etiological agent of cutaneous leishmaniasis in Brazil. Twenty-three compounds were able to inhibit ≥ 80 % L. braziliensis growth at 5 μM. Six out of the PBox selected 23 compounds were found to be highly selective against L. braziliensis intracellular amastigotes with selectivity index varying from > 104 to > 746 and IC50s ranging from 47 to 480 nM. The compounds were also active against antimony-resistant L. braziliensis isolated from the field or laboratory selected mutants, revealing the potential on treating patients infected with drug resistant parasites. Most of the selected compounds were known to be active against kinetoplastids, however, two compounds (MMV688703 and MMV676477) were part of toxoplasmosis and tuberculosis 'PBox' disease set, reinforcing the potential of phenotyping screening to unveil drug repurposing. Here we applied a computational prediction of pharmacokinetic properties using the ADMET predictor pkCSM (http://biosig.unimelb.edu.au/pkcsm/). The tool offered clues on potential drug development needs and can support further in vivo studies. Molecular docking analysis identified CRK3 (LbrM.35.0660), CYP450 (LbrM.30.3580) and PKA (LbrM.18.1180) as L. braziliensis targets for MMV676604, MMV688372 and MMV688703, respectively. Compounds from 'Pathogen Box' thus represents a new hope for novel (or repurposed) small molecules source to tackle leishmaniases.
Collapse
Affiliation(s)
- Juliano A Souza Silva
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-009, MG, Brazil.
| | - Luiza G Tunes
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-009, MG, Brazil.
| | - Roney S Coimbra
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-009, MG, Brazil.
| | - David B Ascher
- Structural Biology and Bioinformatics, Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21 Institute, 30 Flemington Rd, Parkville, VIC 3052, Melbourne, Australia; Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, VIC 3004, Melbourne, Australia.
| | - Douglas E V Pires
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-009, MG, Brazil; School of Computing and Information Systems, The University of Melbourne, Doug McDonell Building, VIC 3010, Parkville, Melbourne, Australia.
| | - Rubens L Monte-Neto
- Instituto René Rachou - Fiocruz Minas, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-009, MG, Brazil.
| |
Collapse
|
23
|
Ang CW, Tan L, Sykes ML, AbuGharbiyeh N, Debnath A, Reid JC, West NP, Avery VM, Cooper MA, Blaskovich MAT. Antitubercular and Antiparasitic 2-Nitroimidazopyrazinones with Improved Potency and Solubility. J Med Chem 2020; 63:15726-15751. [PMID: 33151678 PMCID: PMC7770830 DOI: 10.1021/acs.jmedchem.0c01372] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Following the approval of delamanid and pretomanid as new drugs
to treat drug-resistant tuberculosis, there is now a renewed interest
in bicyclic nitroimidazole scaffolds as a source of therapeutics against
infectious diseases. We recently described a nitroimidazopyrazinone
bicyclic subclass with promising antitubercular and antiparasitic
activity, prompting additional efforts to generate analogs with improved
solubility and enhanced potency. The key pendant aryl substituent
was modified by (i) introducing polar functionality to the methylene
linker, (ii) replacing the terminal phenyl group with less lipophilic
heterocycles, or (iii) generating extended biaryl side chains. Improved
antitubercular and antitrypanosomal activity was observed with the
biaryl side chains, with most analogs achieved 2- to 175-fold higher
activity than the monoaryl parent compounds, with encouraging improvements
in solubility when pyridyl groups were incorporated. This study has
contributed to understanding the existing structure–activity
relationship (SAR) of the nitroimidazopyrazinone scaffold against
a panel of disease-causing organisms to support future lead optimization.
Collapse
Affiliation(s)
- Chee Wei Ang
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Australian Infectious Diseases Research Centre, St. Lucia, Queensland 4067, Australia
| | - Melissa L Sykes
- Discovery Biology, Griffith University, Nathan, Queensland 4111, Australia
| | - Neda AbuGharbiyeh
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - 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 92093, United States
| | - Janet C Reid
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Nicholas P West
- School of Chemistry and Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Australian Infectious Diseases Research Centre, St. Lucia, Queensland 4067, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith University, Nathan, Queensland 4111, Australia
| | - Matthew A Cooper
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Australian Infectious Diseases Research Centre, St. Lucia, Queensland 4067, Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Australian Infectious Diseases Research Centre, St. Lucia, Queensland 4067, Australia
| |
Collapse
|
24
|
Ullah I, Gahalawat S, Booshehri LM, Niederstrasser H, Majumdar S, Leija C, Bradford JM, Hu B, Ready JM, Wetzel DM. An Antiparasitic Compound from the Medicines for Malaria Venture Pathogen Box Promotes Leishmania Tubulin Polymerization. ACS Infect Dis 2020; 6:2057-2072. [PMID: 32686409 PMCID: PMC8059355 DOI: 10.1021/acsinfecdis.0c00122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The few frontline antileishmanial drugs are poorly effective and toxic. To search for new drugs for this neglected tropical disease, we tested the activity of compounds in the Medicines for Malaria Venture (MMV) "Pathogen Box" against Leishmania amazonensis axenic amastigotes. Screening yielded six discovery antileishmanial compounds with EC50 values from 50 to 480 nM. Concentration-response assays demonstrated that the best hit, MMV676477, had mid-nanomolar cytocidal potency against intracellular Leishmania amastigotes, Trypanosoma brucei, and Plasmodium falciparum, suggesting broad antiparasitic activity. We explored structure-activity relationships (SAR) within a small group of MMV676477 analogs and observed a wide potency range (20-5000 nM) against axenic Leishmania amastigotes. Compared to MMV676477, our most potent analog, SW41, had ∼5-fold improved antileishmanial potency. Multiple lines of evidence suggest that MMV676477 selectively disrupts Leishmania tubulin dynamics. Morphological studies indicated that MMV676477 and analogs affected L. amazonensis during cell division. Differential centrifugation showed that MMV676477 promoted partitioning of cellular tubulin toward the polymeric form in parasites. Turbidity assays with purified Leishmania and porcine tubulin demonstrated that MMV676477 promoted leishmanial tubulin polymerization in a concentration-dependent manner. Analogs' antiparasitic activity correlated with their ability to facilitate purified Leishmania tubulin polymerization. Chemical cross-linking demonstrated binding of the MMV676477 scaffold to purified Leishmania tubulin, and competition studies established a correlation between binding and antileishmanial activity. Our studies demonstrate that MMV676477 is a potent antiparasitic compound that preferentially promotes Leishmania microtubule polymerization. Due to its selectivity for and broad-spectrum activity against multiple parasites, this scaffold shows promise for antiparasitic drug development.
Collapse
Affiliation(s)
- Imran Ullah
- Department of Pediatrics and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Laela M. Booshehri
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Hanspeter Niederstrasser
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Shreoshi Majumdar
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Christopher Leija
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - James M. Bradford
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Bin Hu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Joseph M. Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Dawn M. Wetzel
- Department of Pediatrics and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
25
|
Alcântara LM, Ferreira TCS, Fontana V, Chatelain E, Moraes CB, Freitas-Junior LH. A Multi-Species Phenotypic Screening Assay for Leishmaniasis Drug Discovery Shows That Active Compounds Display a High Degree of Species-Specificity. Molecules 2020; 25:E2551. [PMID: 32486239 PMCID: PMC7321149 DOI: 10.3390/molecules25112551] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/23/2020] [Accepted: 05/24/2020] [Indexed: 12/05/2022] Open
Abstract
High genetic and phenotypic variability between Leishmania species and strains within species make the development of broad-spectrum antileishmanial drugs challenging. Thus, screening panels consisting of several diverse Leishmania species can be useful in enabling compound prioritization based on their spectrum of activity. In this study, a robust and reproducible high content assay was developed, and 1280 small molecules were simultaneously screened against clinically relevant cutaneous and visceral species: L. amazonensis, L. braziliensis, and L. donovani. The assay is based on THP-1 macrophages infected with stationary phase promastigotes and posterior evaluation of both compound antileishmanial activity and host cell toxicity. The profile of compound activity was species-specific, and out of 51 active compounds, only 14 presented broad-spectrum activity against the three species, with activities ranging from 52% to 100%. Notably, the compounds CB1954, Clomipramine, Maprotiline, Protriptyline, and ML-9 presented pan-leishmanial activity, with efficacy greater than 70%. The results highlight the reduced number of compound classes with pan-leishmanial activity that might be available from diversity libraries, emphasizing the need to screen active compounds against a panel of species and strains. The assay reported here can be adapted to virtually any Leishmania species without the need for genetic modification of parasites, providing the basis for the discovery of broad spectrum anti-leishmanial agents.
Collapse
Affiliation(s)
- Laura M. Alcântara
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP 13083-970, Brazil; (L.M.A.); (T.C.S.F.); (V.F.)
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-900, Brazil
| | - Thalita C. S. Ferreira
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP 13083-970, Brazil; (L.M.A.); (T.C.S.F.); (V.F.)
- Instituto Butantan, São Paulo, SP 05503-900, Brazil
| | - Vanessa Fontana
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP 13083-970, Brazil; (L.M.A.); (T.C.S.F.); (V.F.)
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative, 1211 Geneva, Switzerland;
| | - Carolina B. Moraes
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP 13083-970, Brazil; (L.M.A.); (T.C.S.F.); (V.F.)
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-900, Brazil
| | - Lucio H. Freitas-Junior
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP 13083-970, Brazil; (L.M.A.); (T.C.S.F.); (V.F.)
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-900, Brazil
- Instituto Butantan, São Paulo, SP 05503-900, Brazil
| |
Collapse
|
26
|
de Souza ML, Gonzaga da Costa LA, Silva EDO, de Sousa ALMD, Dos Santos WM, Rolim Neto PJ. Recent strategies for the development of oral medicines for the treatment of visceral leishmaniasis. Drug Dev Res 2020; 81:803-814. [PMID: 32394440 DOI: 10.1002/ddr.21684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 01/02/2023]
Abstract
Considered prevalent in many countries on five continents, especially in low-income regions, leishmaniasis is a neglected tropical disease classified by World Health Organization as one of the diseases for which the development of new treatments is a priority. It is an infectious disease caused by protozoa of the genus Leishmania, whose species may cause different clinical manifestations, such as cutaneous and visceral leishmaniasis (VL). Treatment is exclusively by drug therapy, as it has not been possible to develop vaccines yet. Currently available drugs are not fully effective in all cases; they have parenteral administration and exhibit a number of serious and very common adverse effects. The only oral drug available is expensive and it is not available in many endemic countries. Injectable administration is the main problem of treatments, since it requires patients to go to health centers, hospitalization and professional administration, which are conditions that are not adapted to the reality of the poverty conditions of patients with the disease. In this context, the development of an oral medicine has become a focus as it may solve many of these issues. Based on this scenario, this review aimed to investigate which therapeutic alternatives have been studied for the development of oral drugs directed to the treatment of human VL.
Collapse
Affiliation(s)
- Myla Lôbo de Souza
- Laboratory of Technology of Medicines, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | | | - Emerson de Oliveira Silva
- Laboratory of Technology of Medicines, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | | | - Widson Michael Dos Santos
- Laboratory of Technology of Medicines, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Pedro José Rolim Neto
- Laboratory of Technology of Medicines, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| |
Collapse
|
27
|
Phan TN, Baek KH, Lee N, Byun SY, Shum D, No JH. In Vitro and in Vivo Activity of mTOR Kinase and PI3K Inhibitors Against Leishmania donovani and Trypanosoma brucei. Molecules 2020; 25:molecules25081980. [PMID: 32340370 PMCID: PMC7221892 DOI: 10.3390/molecules25081980] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/02/2020] [Accepted: 04/21/2020] [Indexed: 11/16/2022] Open
Abstract
Kinetoplastid parasites, including Leishmania and Trypanosoma spp., are life threatening pathogens with a worldwide distribution. Next-generation therapeutics for treatment are needed as current treatments have limitations, such as toxicity and drug resistance. In this study, we examined the activities of established mammalian target of rapamycin (mTOR)/phosphoinositide 3-kinase (PI3K) inhibitors against these tropical diseases. High-throughput screening of a library of 1742 bioactive compounds against intracellular L. donovani was performed, and seven mTOR/PI3K inhibitors were identified. Dose-dilution assays revealed that these inhibitors had half maximal effective concentration (EC50) values ranging from 0.14 to 13.44 μM for L. donovani amastigotes and from 0.00005 to 8.16 μM for T. brucei. The results of a visceral leishmaniasis mouse model indicated that treatment with Torin2, dactolisib, or NVP-BGT226 resulted in reductions of 35%, 53%, and 54%, respectively, in the numbers of liver parasites. In an acute T. brucei mouse model using NVP-BGT226 parasite numbers were reduced to under the limits of detection by five consecutive days of treatment. Multiple sequence and structural alignment results indicated high similarities between mTOR and kinetoplastid TORs; the inhibitors are predicted to bind in a similar manner. Taken together, these results indicated that the TOR pathways of parasites have potential for the discovery of novel targets and new potent inhibitors.
Collapse
Affiliation(s)
- Trong-Nhat Phan
- Leishmania Research Laboratory, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Korea; (T.-N.P.); (K.-H.B.)
| | - Kyung-Hwa Baek
- Leishmania Research Laboratory, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Korea; (T.-N.P.); (K.-H.B.)
| | - Nakyung Lee
- Screening Development Platform, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Korea; (N.L.); (S.Y.B.); (D.S.)
| | - Soo Young Byun
- Screening Development Platform, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Korea; (N.L.); (S.Y.B.); (D.S.)
| | - David Shum
- Screening Development Platform, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Korea; (N.L.); (S.Y.B.); (D.S.)
| | - Joo Hwan No
- Leishmania Research Laboratory, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463–400, Korea; (T.-N.P.); (K.-H.B.)
- Correspondence:
| |
Collapse
|
28
|
Pedron J, Boudot C, Brossas JY, Pinault E, Bourgeade-Delmas S, Sournia-Saquet A, Boutet-Robinet E, Destere A, Tronnet A, Bergé J, Bonduelle C, Deraeve C, Pratviel G, Stigliani JL, Paris L, Mazier D, Corvaisier S, Since M, Malzert-Fréon A, Wyllie S, Milne R, Fairlamb AH, Valentin A, Courtioux B, Verhaeghe P. New 8-Nitroquinolinone Derivative Displaying Submicromolar in Vitro Activities against Both Trypanosoma brucei and cruzi. ACS Med Chem Lett 2020; 11:464-472. [PMID: 32292551 PMCID: PMC7153024 DOI: 10.1021/acsmedchemlett.9b00566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/06/2020] [Indexed: 11/28/2022] Open
Abstract
An antikinetoplastid pharmacomodulation study was conducted at position 6 of the 8-nitroquinolin-2(1H)-one pharmacophore. Fifteen new derivatives were synthesized and evaluated in vitro against L. infantum, T. brucei brucei, and T. cruzi, in parallel with a cytotoxicity assay on the human HepG2 cell line. A potent and selective 6-bromo-substituted antitrypanosomal derivative 12 was revealed, presenting EC50 values of 12 and 500 nM on T. b. brucei trypomastigotes and T. cruzi amastigotes respectively, in comparison with four reference drugs (30 nM ≤ EC50 ≤ 13 μM). Moreover, compound 12 was not genotoxic in the comet assay and showed high in vitro microsomal stability (half life >40 min) as well as favorable pharmacokinetic behavior in the mouse after oral administration. Finally, molecule 12 (E° = -0.37 V/NHE) was shown to be bioactivated by type 1 nitroreductases, in both Leishmania and Trypanosoma, and appears to be a good candidate to search for novel antitrypanosomal lead compounds.
Collapse
Affiliation(s)
- Julien Pedron
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Clotilde Boudot
- Université de Limoges, UMR INSERM 1094, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025 Limoges, France
| | - Jean-Yves Brossas
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Parasitologie Mycologie, 75013 Paris, France
| | - Emilie Pinault
- Université de Limoges, BISCEm Mass Spectrometry Platform, CBRS, 2 rue du Pr. Descottes, F-87025 Limoges, France
| | | | | | - Elisa Boutet-Robinet
- Toxalim, Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, 31077 Toulouse, France
| | - Alexandre Destere
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, France, INSERM, UMR 1248, University of Limoges, F-87025 Limoges, France
| | - Antoine Tronnet
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Justine Bergé
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Colin Bonduelle
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Céline Deraeve
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | | | | | - Luc Paris
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Parasitologie Mycologie, 75013 Paris, France
| | - Dominique Mazier
- CIMI-Paris, Sorbonne Université, 91 Boulevard de l’Hôpital, 75013 Paris, France
| | - Sophie Corvaisier
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Université, 14032 Caen, France
| | - Marc Since
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Université, 14032 Caen, France
| | - Aurélie Malzert-Fréon
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Université, 14032 Caen, France
| | - Susan Wyllie
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Rachel Milne
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Alan H. Fairlamb
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Alexis Valentin
- UMR 152 PharmaDev, Université de Toulouse, IRD, UPS, 31077 Toulouse, France
| | - Bertrand Courtioux
- Université de Limoges, UMR INSERM 1094, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025 Limoges, France
| | - Pierre Verhaeghe
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| |
Collapse
|
29
|
Tadele M, Abay SM, Makonnen E, Hailu A. Leishmania donovani Growth Inhibitors from Pathogen Box Compounds of Medicine for Malaria Venture. Drug Des Devel Ther 2020; 14:1307-1317. [PMID: 32280200 PMCID: PMC7130106 DOI: 10.2147/dddt.s244903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/18/2020] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Leishmaniasis is a collective term used to describe various pathological conditions caused by an obligate intracellular protozoan of the genus Leishmania. It is one of the neglected diseases and has been given minimal attention by drug discovery and development stakeholders to narrow the safety and efficacy gaps of the drugs currently used to treat leishmaniasis. The challenge is further exacerbated by the emergence of drug resistance by the parasites. METHODS Aiming to look for potential anti-leishmanial hits and leads, we screened Medicines for Malaria Venture (MMV) Pathogen Box compounds against clinically isolated Leishmania donovani strain. In this medium-throughput primary screening assay, the compounds were screened against promastigotes, and then against amastigote stages. RESULTS From the total 400 compounds screened, 35 compounds showed >50% inhibitory activity on promastigotes in the initial screen (1 μM). Out of these compounds, nine showed >70% inhibition, with median inhibitory concentration (IC50) ranging from 12 to 491 nM using the anti-promastigote assay, and from 53 to 704 nM using the intracellular amastigote assay. Identified compounds demonstrated acceptable safety profiles on THP-1 cell lines and sheep red blood cells, and had appropriate physicochemical properties suitable for further drug development. Two compounds (MMV690102 and MMV688262) were identified as leads. The anti-tubercular agent MMV688262 (delamanid) showed a synergistic effect with amphotericin B, indicating the prospect of using this compound for combination therapy. CONCLUSION The current study indicates the presence of additional hits which may hold promise as starting points for anti-leishmanial drug discovery and in-depth structure-activity relationship studies.
Collapse
Affiliation(s)
- Markos Tadele
- Animal Health Research Program, Ethiopian Institute of Agricultural Research, Holetta, Ethiopia
| | - Solomon M Abay
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Eyasu Makonnen
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Asrat Hailu
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
30
|
Santos SS, de Araújo RV, Giarolla J, Seoud OE, Ferreira EI. Searching for drugs for Chagas disease, leishmaniasis and schistosomiasis: a review. Int J Antimicrob Agents 2020; 55:105906. [PMID: 31987883 DOI: 10.1016/j.ijantimicag.2020.105906] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
Abstract
Chagas disease, leishmaniasis and schistosomiasis are neglected diseases (NDs) and are a considerable global challenge. Despite the huge number of people infected, NDs do not create interest from pharmaceutical companies because the associated revenue is generally low. Most of the research on these diseases has been conducted in academic institutions. The chemotherapeutic armamentarium for NDs is scarce and inefficient and better drugs are needed. Researchers have found some promising potential drug candidates using medicinal chemistry and computational approaches. Most of these compounds are synthetic but some are from natural sources or are semi-synthetic. Drug repurposing or repositioning has also been greatly stimulated for NDs. This review considers some potential drug candidates and provides details of their design, discovery and activity.
Collapse
Affiliation(s)
- Soraya Silva Santos
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Renan Vinicius de Araújo
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Jeanine Giarolla
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Omar El Seoud
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Elizabeth Igne Ferreira
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil.
| |
Collapse
|
31
|
Naderi M, Lemoine JM, Govindaraj RG, Kana OZ, Feinstein WP, Brylinski M. Binding site matching in rational drug design: algorithms and applications. Brief Bioinform 2019; 20:2167-2184. [PMID: 30169563 PMCID: PMC6954434 DOI: 10.1093/bib/bby078] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/18/2018] [Accepted: 07/29/2018] [Indexed: 01/06/2023] Open
Abstract
Interactions between proteins and small molecules are critical for biological functions. These interactions often occur in small cavities within protein structures, known as ligand-binding pockets. Understanding the physicochemical qualities of binding pockets is essential to improve not only our basic knowledge of biological systems, but also drug development procedures. In order to quantify similarities among pockets in terms of their geometries and chemical properties, either bound ligands can be compared to one another or binding sites can be matched directly. Both perspectives routinely take advantage of computational methods including various techniques to represent and compare small molecules as well as local protein structures. In this review, we survey 12 tools widely used to match pockets. These methods are divided into five categories based on the algorithm implemented to construct binding-site alignments. In addition to the comprehensive analysis of their algorithms, test sets and the performance of each method are described. We also discuss general pharmacological applications of computational pocket matching in drug repurposing, polypharmacology and side effects. Reflecting on the importance of these techniques in drug discovery, in the end, we elaborate on the development of more accurate meta-predictors, the incorporation of protein flexibility and the integration of powerful artificial intelligence technologies such as deep learning.
Collapse
Affiliation(s)
- Misagh Naderi
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Jeffrey Mitchell Lemoine
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Division of Computer Science and Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | - Omar Zade Kana
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Wei Pan Feinstein
- High-Performance Computing, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Computation & Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
32
|
Patterson S, Fairlamb AH. Current and Future Prospects of Nitro-compounds as Drugs for Trypanosomiasis and Leishmaniasis. Curr Med Chem 2019; 26:4454-4475. [PMID: 29701144 DOI: 10.2174/0929867325666180426164352] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/01/2018] [Accepted: 04/13/2018] [Indexed: 01/13/2023]
Abstract
Interest in nitroheterocyclic drugs for the treatment of infectious diseases has undergone a resurgence in recent years. Here we review the current status of monocyclic and bicyclic nitroheterocyclic compounds as existing or potential new treatments for visceral leishmaniasis, Chagas' disease and human African trypanosomiasis. Both monocyclic (nifurtimox, benznidazole and fexinidazole) and bicyclic (pretomanid (PA-824) and delamanid (OPC-67683)) nitro-compounds are prodrugs, requiring enzymatic activation to exert their parasite toxicity. Current understanding of the nitroreductases involved in activation and possible mechanisms by which parasites develop resistance is discussed along with a description of the pharmacokinetic / pharmacodynamic behaviour and chemical structure-activity relationships of drugs and experimental compounds.
Collapse
Affiliation(s)
- Stephen Patterson
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Alan H Fairlamb
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
33
|
Rajoli RKR, Podany AT, Moss DM, Swindells S, Flexner C, Owen A, Siccardi M. Modelling the long-acting administration of anti-tuberculosis agents using PBPK: a proof of concept study. Int J Tuberc Lung Dis 2019; 22:937-944. [PMID: 29991405 DOI: 10.5588/ijtld.17.0515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
SETTING Anti-tuberculosis formulations necessitate uninterrupted treatment to cure tuberculosis (TB), but are characterised by suboptimal adherence, which jeopardises therapeutic efficacy. Long-acting injectable (LAI) formulations or implants could address these associated issues. OBJECTIVE niazid, rifapentine, bedaquiline and delamanid-in adults for treatment for latent tuberculous infection (LTBI). DESIGN PBPK models were developed and qualified against available clinical data by integrating drug physicochemical properties and in vitro and population pharmacokinetic data into a mechanistic description of drug distribution. Combinations of optimal dose and release rates were simulated such that plasma concentrations were maintained over the epidemiological cut-off or minimum inhibitory concentration for the dosing interval. RESULTS The PBPK model identified 1500 mg of delamanid and 250 mg of rifapentine as sufficient doses for monthly intramuscular administration, if a formulation or device can deliver the required release kinetics of 0.001-0.0025 h-1 and 0.0015-0.0025 h-1, respectively. Bedaquiline and isoniazid would require weekly to biweekly intramuscular dosing. CONCLUSION We identified the theoretical doses and release rates of LAI anti-tuberculosis formulations. Such a strategy could ease the problem of suboptimal adherence provided the associated technological complexities for LTBI treatment are addressed.
Collapse
Affiliation(s)
- R K R Rajoli
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - A T Podany
- College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - D M Moss
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK, School of Pharmacy, Keele University, Newcastle, UK
| | - S Swindells
- College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - C Flexner
- Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - A Owen
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - M Siccardi
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| |
Collapse
|
34
|
Need for sustainable approaches in antileishmanial drug discovery. Parasitol Res 2019; 118:2743-2752. [DOI: 10.1007/s00436-019-06443-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
|
35
|
Abstract
Abstract
Leishmaniasis is a group of zoonotic diseases caused by a trypanosomatid parasite mostly in impoverished populations of low-income countries. In their different forms, leishmaniasis is prevalent in more than 98 countries all over the world and approximately 360-million people are at risk. Since no vaccine is currently available to prevent any form of the disease, the control strategy of leishmaniasis mainly relies on early case detection followed by adequate pharmacological treatment that may improve the prognosis and can reduce transmission. A handful of compounds and formulations are available for the treatment of leishmaniasis in humans, but only few of them are currently in use since most of these agents are associated with toxicity problems such as nephrotoxicity and cardiotoxicity in addition to resistance problems. In recent decades, very few novel drugs, new formulations of standard drugs or combinations of them have been approved against leishmaniasis. This review highlights the current drugs and combinations that are used medical practice and recent advances in new treatments against leishmaniasis that were pointed out in the recent 2nd Conference, Global Challenges in Neglected Tropical Diseases, held in San Juan, Puerto Rico in June 2018, emphasizing the plethora of new families of molecules that are bridging the gap between preclinical and first-in-man trials in next future.
Collapse
|
36
|
Sundar S, Agrawal N, Singh B. Exploiting knowledge on pharmacodynamics-pharmacokinetics for accelerated anti-leishmanial drug discovery/development. Expert Opin Drug Metab Toxicol 2019; 15:595-612. [PMID: 31174439 DOI: 10.1080/17425255.2019.1629417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Being on the top list of neglected tropical diseases, leishmaniasis has been marked for elimination by 2020. In the light of small armamentarium of drugs and their associated drawbacks, the understanding of pharmacodynamics and/or pharmacokinetics becomes a priority to achieve and sustain disease elimination. Areas covered: The authors have looked into pharmacological aspects of existing and emerging drugs for treatment of leishmaniasis. An in-depth understanding of pharmacodynamics and pharmacokinetics (PKPD) provides a rationale for drug designing and optimizing the treatment strategies. It forms a key to prevent drug resistance and avoid drug-associated adverse effects. The authors have compiled the researches on the PKPD of different anti-leishmanial formulations that have the potential for improved and/or effective disease intervention. Expert opinion: Understanding the pharmacological aspects of drugs forms the basis for the clinical application of novel drugs. Tailoring drug dosage and individualized treatment can avoid the adverse events and bridge gap between the in vitro models and their clinical application. An integrated approach, with pragmatic use of technological advances can improve phenotypic screening and physiochemical properties of novel drugs. Concomitantly, this can serve to improve clinical efficacies, reduce the incidence of relapse and accelerate the drug discovery/development process for leishmaniasis elimination.
Collapse
Affiliation(s)
- Shyam Sundar
- a Department of Medicine , Institute of Medical Sciences, Banaras Hindu University , Varanasi , India
| | - Neha Agrawal
- b Hepatology , Temple University , Philadelphia , PA , USA
| | - Bhawana Singh
- a Department of Medicine , Institute of Medical Sciences, Banaras Hindu University , Varanasi , India.,c Department of Pathology , Wexner Medical Center, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
37
|
Upadhyay A, Chandrakar P, Gupta S, Parmar N, Singh SK, Rashid M, Kushwaha P, Wahajuddin M, Sashidhara KV, Kar S. Synthesis, Biological Evaluation, Structure-Activity Relationship, and Mechanism of Action Studies of Quinoline-Metronidazole Derivatives Against Experimental Visceral Leishmaniasis. J Med Chem 2019; 62:5655-5671. [PMID: 31124675 DOI: 10.1021/acs.jmedchem.9b00628] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In our efforts to identify novel chemical scaffolds for the development of antileishmanial agents, a series of quinoline-metronidazole hybrid compounds was synthesized and tested against the murine model of visceral leishmaniasis. Among all synthesized derivatives, 15b and 15i showed significant antileishmanial efficacy against both extracellular promastigote (IC50 9.54 and 5.42 μM, respectively) and intracellular amastigote (IC50 9.81 and 3.75 μM, respectively) forms of Leishmania donovani with negligible cytotoxicity toward the host (J774 macrophages, Vero cells). However, compound 15i effectively inhibited the parasite burden in the liver and spleen (>80%) of infected BALB/c mice. Mechanistic studies revealed that 15i triggers oxidative stress which induces bioenergetic collapse and apoptosis of the parasite by decreasing ATP production and mitochondrial membrane potential. Structure-activity analyses and pharmacokinetic studies suggest 15i as a promising antileishmanial lead and emphasize the importance of quinoline-metronidazole series as a suitable platform for the future development of antileishmanial agents.
Collapse
Affiliation(s)
- Akanksha Upadhyay
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Pragya Chandrakar
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Sampa Gupta
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Naveen Parmar
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Sandeep Kumar Singh
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Mamunur Rashid
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Pragati Kushwaha
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Muhammad Wahajuddin
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Koneni V Sashidhara
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| | - Susanta Kar
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan , New Delhi 110025 , India
| |
Collapse
|
38
|
Chakravarty J, Sundar S. Current and emerging medications for the treatment of leishmaniasis. Expert Opin Pharmacother 2019; 20:1251-1265. [DOI: 10.1080/14656566.2019.1609940] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jaya Chakravarty
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
39
|
Gómara M, Ramón-García S. The FICI paradigm: Correcting flaws in antimicrobial in vitro synergy screens at their inception. Biochem Pharmacol 2019; 163:299-307. [PMID: 30836058 DOI: 10.1016/j.bcp.2019.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/01/2019] [Indexed: 10/27/2022]
Abstract
Antibiotics have become the corner stone of modern medicine. However, our society is currently facing one of the greatest challenges of its time: the emergence of antimicrobial resistance. It is estimated that if no new therapies are implemented by 2050, 10 million people will die worldwide every year as a result of infections caused by bacteria resistant to current antibiotics; new antimicrobials are thus urgently needed. However, drug development is a tedious and very costly endeavor of hundreds of millions that can take up to 15-20 years from the bench discovery to the bedside. Under this scenario, drug repurposing, which consists in identifying new uses for old, clinically approved drugs, has gathered momentum within the pharmaceutical industry. Because most of these drugs have safety and toxicity information packages available, clinical evaluation could be done in a much shorter period than standard timelines. Synergistic combinations of these clinically approved drugs could also be a promising approach to identify novel antimicrobial therapies that might provide rational choices of available drugs to shorten treatment, increase efficacy, reduce toxicity, prevent resistance and treat infections caused by drug-resistant strains. However, although simple in its conception, translating results from in vitro synergy screens into in vivo efficacy or the clinical practice has proven to be a paramount challenge. In this Commentary, we will discuss common flaws at the inception of synergy research programs, with a special focus on the use of the Fractional Inhibitory Concentration Index (FICI), and evaluate potential interventions that can be made at different developmental pre-clinical stages in order to improve the odds of translation from in vitro studies.
Collapse
Affiliation(s)
- Marta Gómara
- Mycobacterial Genetics Group, Department of Microbiology, Preventive Medicine and Public Health. Faculty of Medicine, University of Zaragoza, Spain
| | - Santiago Ramón-García
- Mycobacterial Genetics Group, Department of Microbiology, Preventive Medicine and Public Health. Faculty of Medicine, University of Zaragoza, Spain; Research & Development Agency of Aragon (ARAID) Foundation, Spain; CIBER Respiratory Diseases, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
40
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
41
|
Fersing C, Basmaciyan L, Boudot C, Pedron J, Hutter S, Cohen A, Castera-Ducros C, Primas N, Laget M, Casanova M, Bourgeade-Delmas S, Piednoel M, Sournia-Saquet A, Belle Mbou V, Courtioux B, Boutet-Robinet É, Since M, Milne R, Wyllie S, Fairlamb AH, Valentin A, Rathelot P, Verhaeghe P, Vanelle P, Azas N. Nongenotoxic 3-Nitroimidazo[1,2- a]pyridines Are NTR1 Substrates That Display Potent in Vitro Antileishmanial Activity. ACS Med Chem Lett 2019; 10:34-39. [PMID: 30655943 DOI: 10.1021/acsmedchemlett.8b00347] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
Abstract
Twenty nine original 3-nitroimidazo[1,2-a]pyridine derivatives, bearing a phenylthio (or benzylthio) moiety at position 8 of the scaffold, were synthesized. In vitro evaluation highlighted compound 5 as an antiparasitic hit molecule displaying low cytotoxicity for the human HepG2 cell line (CC50 > 100 μM) alongside good antileishmanial activities (IC50 = 1-2.1 μM) against L. donovani, L. infantum, and L. major; and good antitrypanosomal activities (IC50 = 1.3-2.2 μM) against T. brucei brucei and T. cruzi, in comparison to several reference drugs such as miltefosine, fexinidazole, eflornithine, and benznidazole (IC50 = 0.6 to 13.3 μM). Molecule 5, presenting a low reduction potential (E° = -0.63 V), was shown to be selectively bioactivated by the L. donovani type 1 nitroreductase (NTR1). Importantly, molecule 5 was neither mutagenic (negative Ames test), nor genotoxic (negative comet assay), in contrast to many other nitroaromatics. Molecule 5 showed poor microsomal stability; however, its main metabolite (sulfoxide) remained both active and nonmutagenic, making 5 a good candidate for further in vivo studies.
Collapse
Affiliation(s)
- Cyril Fersing
- Aix Marseille Univ, CNRS, ICR UMR 7273, Équipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | | | - Clotilde Boudot
- Université de Limoges, UMR INSERM 1094, Neuroépidémiologie Tropicale, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025 Limoges, France
| | - Julien Pedron
- LCC−CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Anita Cohen
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
| | - Caroline Castera-Ducros
- Aix Marseille Univ, CNRS, ICR UMR 7273, Équipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | - Nicolas Primas
- Aix Marseille Univ, CNRS, ICR UMR 7273, Équipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | - Michèle Laget
- Aix Marseille Univ, INSERM, UMR MD1, U1261,
SSA, MCT, Marseille, France
| | - Magali Casanova
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
| | | | - Mélanie Piednoel
- Aix Marseille Univ, CNRS, ICR UMR 7273, Équipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | | | - Valère Belle Mbou
- CHU de Limoges, Service d’anatomopathologie, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Bertrand Courtioux
- Université de Limoges, UMR INSERM 1094, Neuroépidémiologie Tropicale, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025 Limoges, France
| | - Élisa Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT,
INP-Purpan, UPS, Toulouse, France
| | - Marc Since
- Centre d’Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ., UNICAEN, CERMN, 14000 Caen, France
| | - Rachel Milne
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, Scotland, United Kingdom
| | - Susan Wyllie
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, Scotland, United Kingdom
| | - Alan H. Fairlamb
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee DD1 5EH, Scotland, United Kingdom
| | - Alexis Valentin
- UMR 152 PharmaDev, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Pascal Rathelot
- Aix Marseille Univ, CNRS, ICR UMR 7273, Équipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | | | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Équipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | - Nadine Azas
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, Marseille, France
| |
Collapse
|
42
|
Molecular Basis of the Leishmanicidal Activity of the Antidepressant Sertraline as a Drug Repurposing Candidate. Antimicrob Agents Chemother 2018; 62:AAC.01928-18. [PMID: 30297370 DOI: 10.1128/aac.01928-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
Drug repurposing affords the implementation of new treatments at a moderate cost and under a faster time-scale. Most of the clinical drugs against Leishmania share this origin. The antidepressant sertraline has been successfully assayed in a murine model of visceral leishmaniasis. Nevertheless, sertraline targets in Leishmania were poorly defined. In order to get a detailed insight into the leishmanicidal mechanism of sertraline on Leishmania infantum, unbiased multiplatform metabolomics and transmission electron microscopy were combined with a focused insight into the sertraline effects on the bioenergetics metabolism of the parasite. Sertraline induced respiration uncoupling, a significant decrease of intracellular ATP level, and oxidative stress in L. infantum promastigotes. Metabolomics evidenced an extended metabolic disarray caused by sertraline. This encompasses a remarkable variation of the levels of thiol-redox and polyamine biosynthetic intermediates, as well as a shortage of intracellular amino acids used as metabolic fuel by Leishmania Sertraline killed Leishmania through a multitarget mechanism of action, tackling essential metabolic pathways of the parasite. As such, sertraline is a valuable candidate for visceral leishmaniasis treatment under a drug repurposing strategy.
Collapse
|
43
|
Abstract
The nitro group is considered to be a versatile and unique functional group in medicinal chemistry. Despite a long history of use in therapeutics, the nitro group has toxicity issues and is often categorized as a structural alert or a toxicophore, and evidence related to drugs containing nitro groups is rather contradictory. In general, drugs containing nitro groups have been extensively associated with mutagenicity and genotoxicity. In this context, efforts toward the structure-mutagenicity or structure-genotoxicity relationships have been undertaken. The current Perspective covers various aspects of agents that contain nitro groups, their bioreductive activation mechanisms, their toxicities, and approaches to combat their toxicity issues. In addition, recent advances in the field of anticancer, antitubercular and antiparasitic agents containing nitro groups, along with a patent survey on hypoxia-activated prodrugs containing nitro groups, are also covered.
Collapse
Affiliation(s)
- Kunal Nepali
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| |
Collapse
|
44
|
Fersing C, Boudot C, Pedron J, Hutter S, Primas N, Castera-Ducros C, Bourgeade-Delmas S, Sournia-Saquet A, Moreau A, Cohen A, Stigliani JL, Pratviel G, Crozet MD, Wyllie S, Fairlamb A, Valentin A, Rathelot P, Azas N, Courtioux B, Verhaeghe P, Vanelle P. 8-Aryl-6-chloro-3-nitro-2-(phenylsulfonylmethyl)imidazo[1,2-a]pyridines as potent antitrypanosomatid molecules bioactivated by type 1 nitroreductases. Eur J Med Chem 2018; 157:115-126. [PMID: 30092366 PMCID: PMC7089781 DOI: 10.1016/j.ejmech.2018.07.064] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/26/2018] [Accepted: 07/28/2018] [Indexed: 11/28/2022]
Abstract
Based on a previously identified antileishmanial 6,8-dibromo-3-nitroimidazo[1,2-a]pyridine derivative, a Suzuki-Miyaura coupling reaction at position 8 of the scaffold was studied and optimized from a 8-bromo-6-chloro-3-nitroimidazo[1,2-a]pyridine substrate. Twenty-one original derivatives were prepared, screened in vitro for activity against L. infantum axenic amastigotes and T. brucei brucei trypomastigotes and evaluated for their cytotoxicity on the HepG2 human cell line. Thus, 7 antileishmanial hit compounds were identified, displaying IC50 values in the 1.1-3 μM range. Compounds 13 and 23, the 2 most selective molecules (SI = >18 or >17) were additionally tested on both the promastigote and intramacrophage amastigote stages of L. donovani. The two molecules presented a good activity (IC50 = 1.2-1.3 μM) on the promastigote stage but only molecule 23, bearing a 4-pyridinyl substituent at position 8, was active on the intracellular amastigote stage, with a good IC50 value (2.3 μM), slightly lower than the one of miltefosine (IC50 = 4.3 μM). The antiparasitic screening also revealed 8 antitrypanosomal hit compounds, including 14 and 20, 2 very active (IC50 = 0.04-0.16 μM) and selective (SI = >313 to 550) molecules toward T. brucei brucei, in comparison with drug-candidate fexinidazole (IC50 = 0.6 & SI > 333) or reference drugs suramin and eflornithine (respective IC50 = 0.03 and 13.3 μM). Introducing an aryl moiety at position 8 of the scaffold quite significantly increased the antitrypanosomal activity of the pharmacophore. Antikinetoplastid molecules 13, 14, 20 and 23 were assessed for bioactivation by parasitic nitroreductases (either in L. donovani or in T. brucei brucei), using genetically modified parasite strains that over-express NTRs: all these molecules are substrates of type 1 nitroreductases (NTR1), such as those that are responsible for the bioactivation of fexinidazole. Reduction potentials measured for these 4 hit compounds were higher than that of fexinidazole (-0.83 V), ranging from -0.70 to -0.64 V.
Collapse
Affiliation(s)
- Cyril Fersing
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, FAC PHARM, 27 Boulevard Jean Moulin, CS30064, 13385, Marseille Cedex 05, France
| | - Clotilde Boudot
- Université de Limoges, UMR Inserm 1094, Neuroépidémiologie Tropicale, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025, Limoges, France
| | - Julien Pedron
- LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sébastien Hutter
- IHU Méditerranée Infection, Aix-Marseille Univ, UMR VITROME, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Nicolas Primas
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, FAC PHARM, 27 Boulevard Jean Moulin, CS30064, 13385, Marseille Cedex 05, France
| | - Caroline Castera-Ducros
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, FAC PHARM, 27 Boulevard Jean Moulin, CS30064, 13385, Marseille Cedex 05, France
| | | | | | - Alain Moreau
- LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Anita Cohen
- IHU Méditerranée Infection, Aix-Marseille Univ, UMR VITROME, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | | | | | - Maxime D Crozet
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, FAC PHARM, 27 Boulevard Jean Moulin, CS30064, 13385, Marseille Cedex 05, France
| | - Susan Wyllie
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee, DD1 5EH, Scotland, United Kingdom
| | - Alan Fairlamb
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee, DD1 5EH, Scotland, United Kingdom
| | - Alexis Valentin
- UMR 152 PHARMA-DEV, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Pascal Rathelot
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, FAC PHARM, 27 Boulevard Jean Moulin, CS30064, 13385, Marseille Cedex 05, France
| | - Nadine Azas
- IHU Méditerranée Infection, Aix-Marseille Univ, UMR VITROME, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Bertrand Courtioux
- Université de Limoges, UMR Inserm 1094, Neuroépidémiologie Tropicale, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025, Limoges, France
| | | | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, FAC PHARM, 27 Boulevard Jean Moulin, CS30064, 13385, Marseille Cedex 05, France.
| |
Collapse
|
45
|
Berry SL, Hameed H, Thomason A, Maciej-Hulme ML, Saif Abou-Akkada S, Horrocks P, Price HP. Development of NanoLuc-PEST expressing Leishmania mexicana as a new drug discovery tool for axenic- and intramacrophage-based assays. PLoS Negl Trop Dis 2018; 12:e0006639. [PMID: 30001317 PMCID: PMC6057649 DOI: 10.1371/journal.pntd.0006639] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/24/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
The protozoan parasite Leishmania causes leishmaniasis; a spectrum of diseases of which there are an estimated 1 million new cases each year. Current treatments are toxic, expensive, difficult to administer, and resistance to them is emerging. New therapeutics are urgently needed, however, screening the infective amastigote form of the parasite is challenging. Only certain species can be differentiated into axenic amastigotes, and compound activity against these does not always correlate with efficacy against the parasite in its intracellular niche. Methods used to assess compound efficacy on intracellular amastigotes often rely on microscopy-based assays. These are laborious, require specialist equipment and can only determine parasite burden, not parasite viability. We have addressed this clear need in the anti-leishmanial drug discovery process by producing a transgenic L. mexicana cell line that expresses the luciferase NanoLuc-PEST. We tested the sensitivity and versatility of this transgenic strain, in comparison with strains expressing NanoLuc and the red-shifted firefly luciferase. We then compared the NanoLuc-PEST luciferase to the current methods in both axenic and intramacrophage amastigotes following treatment with a supralethal dose of Amphotericin B. NanoLuc-PEST was a more dynamic indicator of cell viability due to its high turnover rate and high signal:background ratio. This, coupled with its sensitivity in the intramacrophage assay, led us to validate the NanoLuc-PEST expressing cell line using the MMV Pathogen Box in a two-step process: i) identify hits against axenic amastigotes, ii) screen these hits using our bioluminescence-based intramacrophage assay. The data obtained from this highlights the potential of compounds active against M. tuberculosis to be re-purposed for use against Leishmania. Our transgenic L. mexicana cell line is therefore a highly sensitive and dynamic system suitable for Leishmania drug discovery in axenic and intramacrophage amastigote models. The protozoan parasite Leishmania causes a spectrum of diseases collectively known as leishmaniasis. The parasite is transmitted to humans by the bite of its vector, the sand fly, following which the parasite invades host white blood cells, particularly macrophages. Leishmaniasis is classified as a neglected tropical disease, and is endemic in 97 countries. Symptoms of the disease depend on the species of Leishmania. These include skin lesions, destruction of the mucosal membranes, and the visceral form which is usually fatal if untreated. Current therapeutic options for leishmaniasis have a number of associated problems that include toxicity, the development of drug resistance and poor patient compliance due to lengthy and painful treatment regimens. New therapeutics are therefore urgently needed. The ability to screen potential drug candidates requires robust screening assays. Currently, screening the intracellular parasite relies on microscopy-based techniques that require expensive equipment, are time consuming and only detect parasite burden, not viability. By using a transgenic cell line that expresses the NanoLuc-PEST luciferase, we show that we have a parasite-specific viability marker that can be used to measure the efficacy of compounds against the intracellular parasite. We validate the potential of this cell line by screening the MMV Pathogen Box.
Collapse
Affiliation(s)
- Sarah L. Berry
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Hamza Hameed
- Institute for Science and Technology in Medicine, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Anna Thomason
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- Current address: School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Marissa L. Maciej-Hulme
- Radboud University Medical Center, Department of Nephrology, Geert Grooteplein 10, GA Nijmegan, The Netherlands
| | - Somaia Saif Abou-Akkada
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Paul Horrocks
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- Institute for Science and Technology in Medicine, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Helen P. Price
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Liu Y, Matsumoto M, Ishida H, Ohguro K, Yoshitake M, Gupta R, Geiter L, Hafkin J. Delamanid: From discovery to its use for pulmonary multidrug-resistant tuberculosis (MDR-TB). Tuberculosis (Edinb) 2018; 111:20-30. [DOI: 10.1016/j.tube.2018.04.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/09/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
|
47
|
Weng HB, Chen HX, Wang MW. Innovation in neglected tropical disease drug discovery and development. Infect Dis Poverty 2018; 7:67. [PMID: 29950174 PMCID: PMC6022351 DOI: 10.1186/s40249-018-0444-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/23/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Neglected tropical diseases (NTDs) are closely related to poverty and affect over a billion people in developing countries. The unmet treatment needs cause high mortality and disability thereby imposing a huge burden with severe social and economic consequences. Although coordinated by the World Health Organization, various philanthropic organizations, national governments and the pharmaceutical industry have been making efforts in improving the situation, the control of NTDs is still inadequate and extremely difficult today. The lack of safe, effective and affordable medicines is a key contributing factor. This paper reviews the recent advances and some of the challenges that we are facing in the fight against NTDs. MAIN BODY In recent years, a number of innovations have demonstrated propensity to promote drug discovery and development for NTDs. Implementation of multilateral collaborations leads to continued efforts and plays a crucial role in drug discovery. Proactive approaches and advanced technologies are urgently needed in drug innovation for NTDs. However, the control and elimination of NTDs remain a formidable task as it requires persistent international cooperation to make sustainable progresses for a long period of time. Some currently employed strategies were proposed and verified to be successful, which involve both mechanisms of 'Push' which aims at cutting the cost of research and development for industry and 'Pull' which aims at increasing market attractiveness. Coupled to this effort should be the exercise of shared responsibility globally to reduce risks, overcome obstacles and maximize benefits. Since NTDs are closely associated with poverty, it is absolutely essential that the stakeholders take concerted and long-term measures to meet multifaceted challenges by alleviating extreme poverty, strengthening social intervention, adapting climate changes, providing effective monitoring and ensuring timely delivery. CONCLUSIONS The ongoing endeavor at the global scale will ultimately benefit the patients, the countries they are living and, hopefully, the manufacturers who provide new preventive, diagnostic and therapeutic products.
Collapse
Affiliation(s)
- Hong-Bo Weng
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Hai-Xia Chen
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Ming-Wei Wang
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203 China
- The National Center for Drug Screening and the CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), 189 Guoshoujing Road, Pudong New District, Shanghai, 201203 China
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai, 201210 China
| |
Collapse
|
48
|
Pedron J, Boudot C, Hutter S, Bourgeade-Delmas S, Stigliani JL, Sournia-Saquet A, Moreau A, Boutet-Robinet E, Paloque L, Mothes E, Laget M, Vendier L, Pratviel G, Wyllie S, Fairlamb A, Azas N, Courtioux B, Valentin A, Verhaeghe P. Novel 8-nitroquinolin-2(1H)-ones as NTR-bioactivated antikinetoplastid molecules: Synthesis, electrochemical and SAR study. Eur J Med Chem 2018; 155:135-152. [PMID: 29885575 DOI: 10.1016/j.ejmech.2018.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 10/14/2022]
Abstract
To study the antiparasitic 8-nitroquinolin-2(1H)-one pharmacophore, a series of 31 derivatives was synthesized in 1-5 steps and evaluated in vitro against both Leishmania infantum and Trypanosoma brucei brucei. In parallel, the reduction potential of all molecules was measured by cyclic voltammetry. Structure-activity relationships first indicated that antileishmanial activity depends on an intramolecular hydrogen bond (described by X-ray diffraction) between the lactam function and the nitro group, which is responsible for an important shift of the redox potential (+0.3 V in comparison with 8-nitroquinoline). With the assistance of computational chemistry, a set of derivatives presenting a large range of redox potentials (from -1.1 to -0.45 V) was designed and provided a list of suitable molecules to be synthesized and tested. This approach highlighted that, in this series, only substrates with a redox potential above -0.6 V display activity toward L. infantum. Nevertheless, such relation between redox potentials and in vitro antiparasitic activities was not observed in T. b. brucei. Compound 22 is a new hit compound in the series, displaying both antileishmanial and antitrypanosomal activity along with a low cytotoxicity on the human HepG2 cell line. Compound 22 is selectively bioactivated by the type 1 nitroreductases (NTR1) of L. donovani and T. brucei brucei. Moreover, despite being mutagenic in the Ames test, as most of nitroaromatic derivatives, compound 22 was not genotoxic in the comet assay. Preliminary in vitro pharmacokinetic parameters were finally determined and pointed out a good in vitro microsomal stability (half-life > 40 min) and a 92% binding to human albumin.
Collapse
Affiliation(s)
- Julien Pedron
- LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Clotilde Boudot
- Université de Limoges, UMR INSERM 1094, Neuroépidémiologie Tropicale, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025, Limoges, France
| | - Sébastien Hutter
- IHU Méditerranée Infection, équipe VITROME « Vecteurs, Infections Tropicales et Méditerranéennes, 19-21 boulevard Jean Moulin, 13385, Marseille Cedex 05, France
| | | | | | | | - Alain Moreau
- LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Elisa Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Lucie Paloque
- LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Michèle Laget
- UMR MD1, U1261, AMU, INSERM, SSA, IRBA, MCT, Marseille, France
| | - Laure Vendier
- LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Susan Wyllie
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee, DD1 5EH, Scotland, United Kingdom
| | - Alan Fairlamb
- University of Dundee, School of Life Sciences, Division of Biological Chemistry and Drug Discovery, Dow Street, Dundee, DD1 5EH, Scotland, United Kingdom
| | - Nadine Azas
- IHU Méditerranée Infection, équipe VITROME « Vecteurs, Infections Tropicales et Méditerranéennes, 19-21 boulevard Jean Moulin, 13385, Marseille Cedex 05, France
| | - Bertrand Courtioux
- Université de Limoges, UMR INSERM 1094, Neuroépidémiologie Tropicale, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025, Limoges, France
| | - Alexis Valentin
- UMR 152 PharmaDev, Université de Toulouse, IRD, UPS, Toulouse, France
| | | |
Collapse
|
49
|
Abstract
INTRODUCTION Parasitic diseases that pose a threat to human life include leishmaniasis - caused by protozoan parasite Leishmania species. Existing drugs have limitations due to deleterious side effects like teratogenicity, high cost and drug resistance. This calls for the need to have an insight into therapeutic aspects of disease. Areas covered: We have identified different drug targets via. molecular, imuunological, metabolic as well as by system biology approaches. We bring these promising drug targets into light so that they can be explored to their maximum. In an effort to bridge the gaps between existing knowledge and prospects of drug discovery, we have compiled interesting studies on drug targets, thereby paving the way for establishment of better therapeutic aspects. Expert opinion: Advancements in technology shed light on many unexplored pathways. Further probing of well established pathways led to the discovery of new drug targets. This review is a comprehensive report on current and emerging drug targets, with emphasis on several metabolic targets, organellar biochemistry, salvage pathways, epigenetics, kinome and more. Identification of new targets can contribute significantly towards strengthening the pipeline for disease elimination.
Collapse
Affiliation(s)
- Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221 005, UP, India
| | - Bhawana Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221 005, UP, India
| |
Collapse
|
50
|
Saha A, Basu M, Ukil A. Recent advances in understanding Leishmania donovani
infection: The importance of diverse host regulatory pathways. IUBMB Life 2018; 70:593-601. [PMID: 29684241 DOI: 10.1002/iub.1759] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/02/2018] [Indexed: 02/03/2023]
Affiliation(s)
- Amrita Saha
- Department of Biochemistry; University of Calcutta; Kolkata West Bengal India
| | - Moumita Basu
- Department of Biochemistry; University of Calcutta; Kolkata West Bengal India
| | - Anindita Ukil
- Department of Biochemistry; University of Calcutta; Kolkata West Bengal India
| |
Collapse
|