1
|
Kirkman T, Dos Santos Silva C, Tosin M, Bertacine Dias MV. How to Find a Fragment: Methods for Screening and Validation in Fragment-Based Drug Discovery. ChemMedChem 2024; 19:e202400342. [PMID: 39198213 DOI: 10.1002/cmdc.202400342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/01/2024]
Abstract
Fragment-based drug discovery (FBDD) is a crucial strategy for developing new drugs that have been applied to diverse targets, from neglected infectious diseases to cancer. With at least seven drugs already launched to the market, this approach has gained interest in both academics and industry in the last 20 years. FBDD relies on screening small libraries with about 1000-2000 compounds of low molecular weight (about 300 Da) using several biophysical methods. Because of the reduced size of the compounds, the chemical space and diversity can be better explored than large libraries used in high throughput screenings. This review summarises the most common biophysical techniques used in fragment screening and orthogonal validation. We also explore the advantages and drawbacks of the different biophysical techniques and examples of applications and strategies.
Collapse
Affiliation(s)
- Tim Kirkman
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Catharina Dos Santos Silva
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1374, CEP 05508-000, São Paulo, SP, Brazil
| | - Manuela Tosin
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Marcio Vinicius Bertacine Dias
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1374, CEP 05508-000, São Paulo, SP, Brazil
| |
Collapse
|
2
|
LaPlante SR, Coric P, Bouaziz S, França TCC. NMR spectroscopy can help accelerate antiviral drug discovery programs. Microbes Infect 2024; 26:105297. [PMID: 38199267 DOI: 10.1016/j.micinf.2024.105297] [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: 07/04/2023] [Revised: 11/21/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024]
Abstract
Small molecule drugs have an important role to play in combating viral infections, and biophysics support has been central for contributing to the discovery and design of direct acting antivirals. Perhaps one of the most successful biophysical tools for this purpose is NMR spectroscopy when utilized strategically and pragmatically within team workflows and timelines. This report describes some clear examples of how NMR applications contributed to the design of antivirals when combined with medicinal chemistry, biochemistry, X-ray crystallography and computational chemistry. Overall, these multidisciplinary approaches allowed teams to reveal and expose compound physical properties from which design ideas were spawned and tested to achieve the desired successes. Examples are discussed for the discovery of antivirals that target HCV, HIV and SARS-CoV-2.
Collapse
Affiliation(s)
- Steven R LaPlante
- Pasteur Network, INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, Québec, H7V 1B7, Canada; NMX Research and Solutions, Inc., 500 Boulevard Cartier Ouest, Laval, Québec, H7V 5B7, Canada; Université Paris Cité, CNRS, CiTCoM, F-75006, Paris, France.
| | - Pascale Coric
- Université Paris Cité, CNRS, CiTCoM, F-75006, Paris, France
| | - Serge Bouaziz
- Université Paris Cité, CNRS, CiTCoM, F-75006, Paris, France
| | - Tanos C C França
- Pasteur Network, INRS-Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, Québec, H7V 1B7, Canada
| |
Collapse
|
3
|
Abdoul-Latif FM, Oumaskour K, Abdallah N, Ainane A, Houmed Aboubaker I, Merito A, Mohamed H, Ainane T. Overview of Research on Leishmaniasis in Africa: Current Status, Diagnosis, Therapeutics, and Recent Advances Using By-Products of the Sargassaceae Family. Pharmaceuticals (Basel) 2024; 17:523. [PMID: 38675483 PMCID: PMC11054980 DOI: 10.3390/ph17040523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
Leishmaniasis in Africa, which has been designated as a priority neglected tropical disease by various global organizations, exerts its impact on millions of individuals, primarily concentrated within this particular region of the world. As a result of the progressively grave epidemiological data, numerous governmental sectors and civil organizations have concentrated their endeavors on this widespread outbreak with the objective of devising appropriate remedies. This comprehensive examination delves into multiple facets of this parasitic ailment, scrutinizing the associated perils, diagnostic intricacies, and deficiencies within the existing therapeutic protocols. Despite the established efficacy of current treatments, they are not immune to deleterious incidents, particularly concerning toxicity and the emergence of parasitic resistance, thus accentuating the necessity of exploring alternative avenues. Consequently, this research not only encompasses conventional therapeutic approaches, but also extends its scope to encompass complementary and alternative medicinal techniques, thereby striving to identify innovative solutions. A particularly auspicious dimension of this study lies in the exploration of natural substances and by-products derived from some brown algae of the Sargassaceae family. These resources possess the potential to assume a pivotal role in the management of leishmaniasis.
Collapse
Affiliation(s)
- Fatouma Mohamed Abdoul-Latif
- Institut Supérieur des Sciences de la Santé (ISSS), Djibouti City P.O. Box 2530, Djibouti
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti City P.O. Box 486, Djibouti
| | - Khadija Oumaskour
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco
| | - Nadira Abdallah
- Institut Supérieur des Sciences de la Santé (ISSS), Djibouti City P.O. Box 2530, Djibouti
| | - Ayoub Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco
| | | | - Ali Merito
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti City P.O. Box 486, Djibouti
| | - Houda Mohamed
- Peltier Hospital of Djibouti, Djibouti City P.O. Box 2123, Djibouti
| | - Tarik Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco
| |
Collapse
|
4
|
Sow AA, Pahmeier F, Ayotte Y, Anton A, Mazeaud C, Charpentier T, Angelo L, Woo S, Cerikan B, Falzarano D, Abrahamyan L, Lamarre A, Labonté P, Cortese M, Bartenschlager R, LaPlante SR, Chatel-Chaix L. N-Phenylpyridine-3-Carboxamide and 6-Acetyl-1H-Indazole Inhibit the RNA Replication Step of the Dengue Virus Life Cycle. Antimicrob Agents Chemother 2023; 67:e0133122. [PMID: 36700643 PMCID: PMC9933715 DOI: 10.1128/aac.01331-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/26/2022] [Indexed: 01/27/2023] Open
Abstract
Dengue virus (DENV) is a Flavivirus that causes the most prevalent arthropod-borne viral disease. Clinical manifestation of DENV infection ranges from asymptomatic to severe symptoms that can lead to death. Unfortunately, no antiviral treatments against DENV are currently available. In order to identify novel DENV inhibitors, we screened a library of 1,604 chemically diversified fragment-based compounds using DENV reporter viruses that allowed quantification of viral replication in infected cells. Following a validation screening, the two best inhibitor candidates were N-phenylpyridine-3-carboxamide (NPP3C) and 6-acetyl-1H-indazole (6A1HI). The half maximal effective concentration of NPP3C and 6A1H1 against DENV were 7.1 μM and 6.5 μM, respectively. 6A1H1 decreased infectious DENV particle production up to 1,000-fold without any cytotoxicity at the used concentrations. While 6A1HI was DENV-specific, NPP3C also inhibited the replication of other flaviviruses such as West Nile virus and Zika virus. Structure-activity relationship (SAR) studies with 151 analogues revealed key structural elements of NPP3C and 6A1HI required for their antiviral activity. Time-of-drug-addition experiments identified a postentry step as a target of these compounds. Consistently, using a DENV subgenomic replicon, we demonstrated that these compounds specifically impede the viral RNA replication step and exhibit a high genetic barrier-to-resistance. In contrast, viral RNA translation and the de novo biogenesis of DENV replication organelles were not affected. Overall, our data unveil NPP3C and 6A1H1 as novel DENV inhibitors. The information revealed by our SAR studies will help chemically optimize NPP3C and 6A1H1 in order to improve their anti-flaviviral potency and to challenge them in in vivo models.
Collapse
Affiliation(s)
- Aïssatou Aïcha Sow
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Felix Pahmeier
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg University, Heidelberg, Germany
| | - Yann Ayotte
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Anaïs Anton
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Clément Mazeaud
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Tania Charpentier
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Léna Angelo
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Simon Woo
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Berati Cerikan
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg University, Heidelberg, Germany
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Levon Abrahamyan
- Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Alain Lamarre
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Patrick Labonté
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Mirko Cortese
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg University, Heidelberg, Germany
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg partner site, Heidelberg, Germany
| | - Steven R. LaPlante
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Laurent Chatel-Chaix
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
- Center of Excellence in Orphan Diseases Research-Foundation Courtois, Quebec, Canada
- Réseau Intersectoriel de Recherche en Santé de l’Université du Québec, Quebec, Canada
| |
Collapse
|
5
|
Fragment-Based Lead Discovery Strategies in Antimicrobial Drug Discovery. Antibiotics (Basel) 2023; 12:antibiotics12020315. [PMID: 36830226 PMCID: PMC9951956 DOI: 10.3390/antibiotics12020315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Fragment-based lead discovery (FBLD) is a powerful application for developing ligands as modulators of disease targets. This approach strategy involves identification of interactions between low-molecular weight compounds (100-300 Da) and their putative targets, often with low affinity (KD ~0.1-1 mM) interactions. The focus of this screening methodology is to optimize and streamline identification of fragments with higher ligand efficiency (LE) than typical high-throughput screening. The focus of this review is on the last half decade of fragment-based drug discovery strategies that have been used for antimicrobial drug discovery.
Collapse
|
6
|
Mollineda-Diogo N, Chaviano-Montes de Oca CS, Sifontes-Rodríguez S, Espinosa-Buitrago T, Monzote-Fidalgo L, Meneses-Marcel A, Morales-Helguera A, Perez-Castillo Y, Arán-Redó V. Antileishmanial activity of 5-nitroindazole derivatives. Ther Adv Infect Dis 2023; 10:20499361231208294. [PMID: 37915499 PMCID: PMC10617274 DOI: 10.1177/20499361231208294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/01/2023] [Indexed: 11/03/2023] Open
Abstract
Background Currently, there is no safe and effective vaccine against leishmaniasis and existing therapies are inadequate due to high toxicity, cost and decreased efficacy caused by the emergence of resistant parasite strains. Some indazole derivatives have shown in vitro and in vivo activity against Trichomonas vaginalis and Trypanosoma cruzi. On that basis, 20 indazole derivatives were tested in vitro against Leishmania amazonensis. Objective To evaluate the in vitro activity of twenty 2-benzyl-5-nitroindazolin-3-one derivatives against L. amazonensis. Design For the selection of promising compounds, it is necessary to evaluate the indicators for in vitro activity. For this aim, a battery of studies for antileishmanial activity and cytotoxicity were implemented. These results enabled the determination of the substituents in the indazole derivatives responsible for activity and selectivity, through the analysis of the structure-activity relationship (SAR). Methods In vitro cytotoxicity against mouse peritoneal macrophages and growth inhibitory activity in promastigotes were evaluated for 20 compounds. Compounds that showed adequate selectivity were tested against intracellular amastigotes. The SAR from the results in promastigotes was represented using the SARANEA software. Results Eight compounds showed selectivity index >10% and 50% inhibitory concentration <1 µM against the promastigote stage. Against intracellular amastigotes, four were as active as Amphotericin B. The best results were obtained for 2-(benzyl-2,3-dihydro-5-nitro-3-oxoindazol-1-yl) ethyl acetate, with 50% inhibitory concentration of 0.46 ± 0.01 µM against amastigotes and a selectivity index of 875. The SAR study showed the positive effect on the selectivity of the hydrophilic fragments substituted in position 1 of 2-benzyl-5- nitroindazolin-3-one, which played a key role in improving the selectivity profile of this series of compounds. Conclusion 2-bencyl-5-nitroindazolin-3-one derivatives showed selective and potent in vitro activity, supporting further investigations on this family of compounds as potential antileishmanial hits.
Collapse
Affiliation(s)
- Niurka Mollineda-Diogo
- Universidad Central “Marta Abreu” de Las Villas, Centro de Bioactivos Químicos, Carretera a Camajuaní Km. 5 ½, Santa Clara, Villa Clara, Cuba
| | | | - Sergio Sifontes-Rodríguez
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Teresa Espinosa-Buitrago
- Facultad de Farmacia, Departamento de Parasitología, Universidad Complutense de Madrid, Madrid, España
| | - Lianet Monzote-Fidalgo
- Instituto de Medicina Tropical “Pedro Kourí” (IPK), Departamento de Parasitología, La Habana, Cuba
| | - Alfredo Meneses-Marcel
- Universidad Central “Marta Abreu” de Las Villas, Centro de Bioactivos Químicos, Villa Clara, Cuba
| | - Aliuska Morales-Helguera
- Universidad Central “Marta Abreu” de Las Villas, Centro de Bioactivos Químicos, Villa Clara, Cuba
| | - Yunierkis Perez-Castillo
- Grupo de Bio-Quimioinformática and Área de Ciencias Aplicadas, Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, CP 170125, Ecuador
| | - Vicente Arán-Redó
- Instituto de Química Médica del Consejo Superior de Investigaciones Científicas de España, Madrid, España
| |
Collapse
|
7
|
Ayotte Y, Bernet E, Bilodeau F, Cimino M, Gagnon D, Lebughe M, Mistretta M, Ogadinma P, Ouali SL, Sow AA, Chatel-Chaix L, Descoteaux A, Manina G, Richard D, Veyrier F, LaPlante SR. Fragment-Based Phenotypic Lead Discovery To Identify New Drug Seeds That Target Infectious Diseases. ACS Chem Biol 2021; 16:2158-2163. [PMID: 34699722 DOI: 10.1021/acschembio.1c00657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fragment-based lead discovery has emerged over the last decades as one of the most powerful techniques for identifying starting chemical matter to target specific proteins or nucleic acids in vitro. However, the use of such low-molecular-weight fragment molecules in cell-based phenotypic assays has been historically avoided because of concerns that bioassays would be insufficiently sensitive to detect the limited potency expected for such small molecules and that the high concentrations required would likely implicate undesirable artifacts. Herein, we applied phenotype cell-based screens using a curated fragment library to identify inhibitors against a range of pathogens including Leishmania, Plasmodium falciparum, Neisseria, Mycobacterium, and flaviviruses. This proof-of-concept shows that fragment-based phenotypic lead discovery (FPLD) can serve as a promising complementary approach for tackling infectious diseases and other drug-discovery programs.
Collapse
Affiliation(s)
- Yann Ayotte
- Institut national de la recherche scientifique - Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Eve Bernet
- Bacterial Symbionts Evolution, Institut national de la recherche scientifique, Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - François Bilodeau
- NMX Research and Solutions, Inc., 500 Boulevard Cartier Ouest, Laval, Quebec H7V 5B7, Canada
| | - Mena Cimino
- Microbial Individuality and Infection Group, Cell Biology and Infection Department, Institut Pasteur, 25-28 Rue du Docteur Roux 75015, Paris, France
| | - Dominic Gagnon
- Centre de recherche du CHU de Québec-Université Laval, Département de Microbiologie-Infectiologie et d’Immunologie, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Marthe Lebughe
- Bacterial Symbionts Evolution, Institut national de la recherche scientifique, Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Maxime Mistretta
- Microbial Individuality and Infection Group, Cell Biology and Infection Department, Institut Pasteur, 25-28 Rue du Docteur Roux 75015, Paris, France
| | - Paul Ogadinma
- NMX Research and Solutions, Inc., 500 Boulevard Cartier Ouest, Laval, Quebec H7V 5B7, Canada
| | - Sarah-Lisa Ouali
- Institut national de la recherche scientifique - Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Aïssatou Aïcha Sow
- Institut national de la recherche scientifique - Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Laurent Chatel-Chaix
- Institut national de la recherche scientifique - Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Albert Descoteaux
- Institut national de la recherche scientifique - Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Giulia Manina
- Microbial Individuality and Infection Group, Cell Biology and Infection Department, Institut Pasteur, 25-28 Rue du Docteur Roux 75015, Paris, France
| | - Dave Richard
- Centre de recherche du CHU de Québec-Université Laval, Département de Microbiologie-Infectiologie et d’Immunologie, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Frédéric Veyrier
- Bacterial Symbionts Evolution, Institut national de la recherche scientifique, Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Steven R. LaPlante
- Institut national de la recherche scientifique - Armand-Frappier Santé Biotechnologie Research Centre, 531 Boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| |
Collapse
|
8
|
Xu M, Zhao C, Zhu B, Wang L, Zhou H, Yan D, Gu Q, Xu J. Discovering High Potent Hsp90 Inhibitors as Antinasopharyngeal Carcinoma Agents through Fragment Assembling Approach. J Med Chem 2021; 64:2010-2023. [PMID: 33543615 DOI: 10.1021/acs.jmedchem.0c01521] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hsp90 is a new promising target for cancer treatment. Many inhibitors have been discovered as therapeutic agents, and some have passed Phase I and II. However, no one is approved by FDA yet. Novel and druggable Hsp90 inhibitors are still demanding. Here, we report a new way to discover high potent Hsp90 inhibitors as antinasopharyngeal carcinoma agents through assembling fragments. With chemotyping analysis, we extract seven chemotypes from 3482 known Hsp90 inhibitors, screen 500 fragments that are compatible with the chemotypes, and confirm 15 anti-Hsp90 fragments. Click chemistry is employed to construct 172 molecules and synthesize 21 compounds among them. The best inhibitor 3d was further optimized and resulted in more potent 4f (IC50 = 0.16 μM). In vitro and in vivo experiments confirmed that 4f is a promising agent against nasopharyngeal carcinoma. This study may provide a strategy in discovering new ligands against targets without well-understood structures.
Collapse
Affiliation(s)
- Mengyang Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Chao Zhao
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- Shenzhen Cell Inspire Therapeutics Co., Ltd., Shenzhen 518101, China
| | - Biying Zhu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Liangyue Wang
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Huihao Zhou
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Daoguang Yan
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- School of Biotechnology and Health Sciences, Wuyi University, 99 Yingbin Road, Jiangmen 529020, China
| |
Collapse
|
9
|
Sodium Tetraphenylborate Displays Selective Bactericidal Activity against Neisseria meningitidis and N. gonorrhoeae and Is Effective at Reducing Bacterial Infection Load. Antimicrob Agents Chemother 2021; 65:AAC.00254-20. [PMID: 33168608 PMCID: PMC7848997 DOI: 10.1128/aac.00254-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 10/30/2020] [Indexed: 11/20/2022] Open
Abstract
Neisseria meningitidis and Neisseria gonorrhoeae, two highly related species that might have emerged from a common commensal ancestor, constitute major human threats. Vaccines are available to prevent N. meningitidis infection, whereas there are only a limited number of antibiotics available for N. gonorrhoeae Unfortunately, some strains of these species are rapidly evolving and capable of escaping human interventions. Thus, it is now urgent to develop new avenues to fight these bacteria. This study reports that a boron-based salt, sodium tetraphenylborate (NaBPh4), displays high bactericidal activity and remarkable specificity against N. meningitidis and N. gonorrhoeae Other closely related commensal species such as Neisseria lactamica, which is found in the normal flora of healthy individuals, were found to be less affected even at 5-fold higher doses of NaBPh4 This specificity was further observed when much lower sensitivity was found for more distant Neisseriaceae species (such as Neisseria elongata or Kingella oralis) and completely unrelated species. Significant boron uptake by N. meningitidis cells was observed after incubation with 5 μM NaBPh4, as measured by inductively coupled plasma mass spectrometry, suggesting that this drug candidate's target(s) could be located intracellularly or within the cell envelope. Furthermore, mutants with slightly decreased susceptibility displayed alterations in genes coding for cell envelope elements, which reduced their virulence in an animal model of infection. Finally, a single dose of NaBPh4 resulted in a significant reduction in bacterial burden in a mouse model of N. meningitidis bacteremia. Although numerous boron-containing species were previously reported for their complex biological activities, the observation of this narrow selectivity is unprecedented and of potential importance from a therapeutic standpoint.
Collapse
|
10
|
Erlanson DA, de Esch IJP, Jahnke W, Johnson CN, Mortenson PN. Fragment-to-Lead Medicinal Chemistry Publications in 2018. J Med Chem 2020; 63:4430-4444. [PMID: 31913033 DOI: 10.1021/acs.jmedchem.9b01581] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This Perspective, the fourth in an annual series, summarizes fragment-to-lead (F2L) success stories published during 2018. Topics such as target class, screening methods, physicochemical properties, and ligand efficiency are discussed for the 2018 examples as well as for the combined 111 F2L examples covering 2015-2018. While the overall properties of fragments and leads have remained constant, a number of new trends are noted, for example, broadening of target class coverage and application of FBDD to covalent inhibitors. Moreover, several studies make use of fragment hits that were previously described in the literature, illustrating that fragments are versatile starting points that can be optimized to structurally diverse leads. By focusing on success stories, the hope is that this Perspective will identify and inform best practices in fragment-based drug discovery.
Collapse
Affiliation(s)
- Daniel A Erlanson
- Frontier Medicines, 151 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Wolfgang Jahnke
- Novartis Institutes for Biomedical Research, Chemical Biology and Therapeutics, 4002 Basel, Switzerland
| | - Christopher N Johnson
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, United Kingdom
| | - Paul N Mortenson
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, United Kingdom
| |
Collapse
|
11
|
Precision medicine review: rare driver mutations and their biophysical classification. Biophys Rev 2019; 11:5-19. [PMID: 30610579 PMCID: PMC6381362 DOI: 10.1007/s12551-018-0496-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023] Open
Abstract
How can biophysical principles help precision medicine identify rare driver mutations? A major tenet of pragmatic approaches to precision oncology and pharmacology is that driver mutations are very frequent. However, frequency is a statistical attribute, not a mechanistic one. Rare mutations can also act through the same mechanism, and as we discuss below, “latent driver” mutations may also follow the same route, with “helper” mutations. Here, we review how biophysics provides mechanistic guidelines that extend precision medicine. We outline principles and strategies, especially focusing on mutations that drive cancer. Biophysics has contributed profoundly to deciphering biological processes. However, driven by data science, precision medicine has skirted some of its major tenets. Data science embodies genomics, tissue- and cell-specific expression levels, making it capable of defining genome- and systems-wide molecular disease signatures. It classifies cancer driver genes/mutations and affected pathways, and its associated protein structural data guide drug discovery. Biophysics complements data science. It considers structures and their heterogeneous ensembles, explains how mutational variants can signal through distinct pathways, and how allo-network drugs can be harnessed. Biophysics clarifies how one mutation—frequent or rare—can affect multiple phenotypic traits by populating conformations that favor interactions with other network modules. It also suggests how to identify such mutations and their signaling consequences. Biophysics offers principles and strategies that can help precision medicine push the boundaries to transform our insight into biological processes and the practice of personalized medicine. By contrast, “phenotypic drug discovery,” which capitalizes on physiological cellular conditions and first-in-class drug discovery, may not capture the proper molecular variant. This is because variants of the same protein can express more than one phenotype, and a phenotype can be encoded by several variants.
Collapse
|
12
|
Schulze J, Baukmann H, Wawrzinek R, Fuchsberger FF, Specker E, Aretz J, Nazaré M, Rademacher C. CellFy: A Cell-Based Fragment Screen against C-Type Lectins. ACS Chem Biol 2018; 13:3229-3235. [PMID: 30480432 DOI: 10.1021/acschembio.8b00875] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Fragment-based drug discovery is a powerful complement to conventional high-throughput screening, especially for difficult targets. Screening low-molecular-weight fragments usually requires highly sensitive biophysical methods, because of the generally low affinity of the identified ligands. Here, we developed a cell-based fragment screening assay (cellFy) that allows sensitive identification of fragment hits in a physiologically more relevant environment, in contrast to isolated target screenings in solution. For this, a fluorescently labeled multivalent reporter was employed, enabling direct measurement of displacement by low-molecular-weight fragments without requiring enzymatic reactions or receptor activation. We applied this technique to identify hits against two challenging targets of the C-type lectin receptor (CLR) family: Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Nonintegrin (DC-SIGN) and Langerin. Both receptors are involved in pathogen recognition and initiation of an immune response, which renders them attractive targets for immune modulation. Because of their shallow and hydrophilic primary binding site, hit identification for CLRs is challenging and druglike ligands for CLRs are sparse. Screening of a fragment library followed by hit validation identified several promising candidates for further fragment evolution for DC-SIGN. In addition, a multiplexed assay format was developed for simultaneous screening against multiple CLRs, allowing a selectivity counterscreening. Overall, this sensitive cell-based fragment screening assay provides a powerful tool for rapid identification of bioactive fragments, even for difficult targets.
Collapse
Affiliation(s)
- Jessica Schulze
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Hannes Baukmann
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Robert Wawrzinek
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Felix F. Fuchsberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Edgar Specker
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Jonas Aretz
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|