1
|
Schultz PG. Synthesis at the Interface of Chemistry and Biology. Acc Chem Res 2024; 57:2631-2642. [PMID: 39198974 PMCID: PMC11443489 DOI: 10.1021/acs.accounts.4c00320] [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: 05/28/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024]
Abstract
ConspectusChemical synthesis as a tool to control the structure and properties of matter is at the heart of chemistry─from the synthesis of fine chemicals and polymers to drugs and solid-state materials. But as the field evolves to tackle larger and larger molecules and molecular complexes, the traditional tools of synthetic chemistry become limiting. In contrast, Mother Nature has developed very different strategies to create the macromolecules and molecular systems that make up the living cell. Our focus has been to ask whether we can use the synthetic strategies and machinery of Mother Nature, together with modern chemical tools, to create new macromolecules, and even whole organisms with properties not existing in nature. One such example involves reprogramming the complex, multicomponent machinery of ribosomal protein synthesis to add new building blocks to the genetic code, overcoming a billion-year constraint on the chemical nature of proteins. This methodology exploits the concept of bioorthogonality to add unique codons, tRNAs and aminoacyl-tRNA synthetases to cells to encode amino acids with physical, chemical and biological properties not found in nature. As a result, we can make precise changes to the structures of proteins, much like those made by chemists to small molecules and beyond those possible by biological approaches alone. This technology has made it possible to probe protein structure and function in vitro and in vivo in ways heretofore not possible, and to make therapeutic proteins with enhanced pharmacology. A second example involves exploiting the molecular diversity of the humoral immune system together with synthetic transition state analogues to make catalytic antibodies, and then expanding this diversity-based strategy (new to chemists at the time) to drug discovery and materials science. This work ushered in a new nature-inspired synthetic strategy in which large libraries of natural or synthetic molecules are designed and then rationally selected or screened for new function, increasing the efficiency by which we can explore chemical space for new physical, chemical and biological properties. A final example is the use of large chemical libraries, robotics and high throughput phenotypic cellular screens to identify small synthetic molecules that can be used to probe and manipulate the complex biology of the cell, exemplified by druglike molecules that control cell fate. This approach provides new insights into complex biology that complements genomic approaches and can lead to new drugs that act by novel mechanisms of action, for example to selectively regenerate tissues. These and other advances have been made possible by using our knowledge of molecular structure and reactivity hand in hand with our understanding of and ability to manipulate the complex machinery of living cells, opening a new frontier in synthesis. This Account overviews the work in my lab and with our collaborators, from our early days to the present, that revolves around this central theme.
Collapse
Affiliation(s)
- Peter G. Schultz
- Department of Chemistry,
L.S. Sam Skaggs Presidential Chair, Scripps
Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
2
|
Zheng W, Lv G, Li J, Zhang Y, Qi W, Yan M, Wu J, Chan C, Pan X, Zhang W. Repurposing of a library for high-content screening of inhibitors against Echinococcus granulosus. Parasit Vectors 2024; 17:373. [PMID: 39227942 PMCID: PMC11370232 DOI: 10.1186/s13071-024-06456-6] [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: 05/08/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Cystic echinococcosis (CE) is a zoonotic disease caused by the larval stage of the dog tapeworm Echinococcus granulosus sensu lato (E. granulosus), with a worldwide distribution. The current treatment strategy for CE is insufficient. Limited drug screening models severely hamper the discovery of effective anti-echinococcosis drugs. METHODS In the present study, using high-content screening technology, we developed a novel high-throughput screening (HTS) assay by counting the ratio of propidium iodide-stained dead protoscoleces (PSCs) to the total number of PSCs. In vitro and ex vivo cyst viability assays were utilized to determine the effect of drugs on cyst viability. RESULTS Using the newly established HTS assay, we screened approximately 12,000 clinical-stage or The Food and Drug Administration (FDA)-approved small molecules from the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library, as well as the LOPAC1280 and SelleckChem libraries, as a strategic approach to facilitate the drug discovery process. Initial screening yielded 173 compounds with anti-echinococcal properties, 52 of which demonstrated dose-response efficacy against E. granulosus PSCs in vitro. Notably, two agents, omaveloxolone and niclosamide, showed complete inhibition upon further validation in cyst and microcyst viability assays in vitro after incubation for 3 days, and in an ex vivo cyst viability assay using cysts isolated from the livers of mice infected with E. granulosus, as determined by morphological assessment. CONCLUSIONS Through the development of a novel HTS assay and by repurposing libraries, we identified omaveloxolone and niclosamide as potent inhibitors against E. granulosus. These compounds show promise as potential anti-echinococcal drugs, and our strategic approach has the potential to promote drug discovery for parasitic infections.
Collapse
Affiliation(s)
- Weinan Zheng
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China.
| | - Gaofei Lv
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China
| | - Jun Li
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yao Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Wenjing Qi
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Mingzhi Yan
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Jinzhi Wu
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China
| | - Chikin Chan
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China
| | - Xiaoben Pan
- Department of Disease Biology, Global Health Drug Discovery Institute, Beijing, 100000, China.
| | - Wenbao Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
3
|
Lenière AC, Vlandas A, Follet J. Treating cryptosporidiosis: A review on drug discovery strategies. Int J Parasitol Drugs Drug Resist 2024; 25:100542. [PMID: 38669849 PMCID: PMC11066572 DOI: 10.1016/j.ijpddr.2024.100542] [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: 11/28/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Despite several decades of research on therapeutics, cryptosporidiosis remains a major concern for human and animal health. Even though this field of research to assess antiparasitic drug activity is highly active and competitive, only one molecule is authorized to be used in humans. However, this molecule was not efficacious in immunocompromised people and the lack of animal therapeutics remains a cause of concern. Indeed, the therapeutic arsenal needs to be developed for both humans and animals. Our work aims to clarify research strategies that historically were diffuse and poorly directed. This paper reviews in vitro and in vivo methodologies to assess the activity of future therapeutic compounds by screening drug libraries or through drug repurposing. It focuses on High Throughput Screening methodologies (HTS) and discusses the lack of knowledge of target mechanisms. In addition, an overview of several specific metabolic pathways and enzymatic activities used as targets against Cryptosporidium is provided. These metabolic processes include glycolytic pathways, fatty acid production, kinase activities, tRNA elaboration, nucleotide synthesis, gene expression and mRNA maturation. As a conclusion, we highlight emerging future strategies for screening natural compounds and assessing drug resistance issues.
Collapse
Affiliation(s)
- Anne-Charlotte Lenière
- University of Lille, CNRS, Centrale Lille, Junia, Université Polytechnique Hauts de France, UMR 8520, IEMN Institut d'Electronique de Microélectronique et de Nanotechnologie, F, 59000, Lille, France
| | - Alexis Vlandas
- University of Lille, CNRS, Centrale Lille, Junia, Université Polytechnique Hauts de France, UMR 8520, IEMN Institut d'Electronique de Microélectronique et de Nanotechnologie, F, 59000, Lille, France
| | - Jérôme Follet
- University of Lille, CNRS, Centrale Lille, Junia, Université Polytechnique Hauts de France, UMR 8520, IEMN Institut d'Electronique de Microélectronique et de Nanotechnologie, F, 59000, Lille, France.
| |
Collapse
|
4
|
Chatterjee AK. Discovery of novel anti-infective agents. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:39-117. [PMID: 39034055 DOI: 10.1016/bs.apha.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Academic and other non-profit institutions have a long-term vision to improve human health where commercial interests can be limited for profit organizations. Medicinal chemistry to these diseases with no commercial benefit needs is well suited in the academic environment and this chapter outlines some work conducted at Calibr-Skaggs around antibiotic drug development that has led to initiation of multiple clinical trials over the last decade.
Collapse
Affiliation(s)
- Arnab K Chatterjee
- Calibr-Skaggs Institute of Innovation Medicines, Scripps Research, La Jolla, CA, United States.
| |
Collapse
|
5
|
Zak J, Pratumchai I, Marro BS, Marquardt KL, Zavareh RB, Lairson LL, Oldstone MBA, Varner JA, Hegerova L, Cao Q, Farooq U, Kenkre VP, Bachanova V, Teijaro JR. JAK inhibition enhances checkpoint blockade immunotherapy in patients with Hodgkin lymphoma. Science 2024; 384:eade8520. [PMID: 38900864 PMCID: PMC11283877 DOI: 10.1126/science.ade8520] [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: 09/21/2022] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
Unleashing antitumor T cell activity by checkpoint inhibitor immunotherapy is effective in cancer patients, but clinical responses are limited. Cytokine signaling through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway correlates with checkpoint immunotherapy resistance. We report a phase I clinical trial of the JAK inhibitor ruxolitinib with anti-PD-1 antibody nivolumab in Hodgkin lymphoma patients relapsed or refractory following checkpoint inhibitor immunotherapy. The combination yielded a best overall response rate of 53% (10/19). Ruxolitinib significantly reduced neutrophil-to-lymphocyte ratios and percentages of myeloid suppressor cells but increased numbers of cytokine-producing T cells. Ruxolitinib rescued the function of exhausted T cells and enhanced the efficacy of immune checkpoint blockade in preclinical solid tumor and lymphoma models. This synergy was characterized by a switch from suppressive to immunostimulatory myeloid cells, which enhanced T cell division.
Collapse
Affiliation(s)
- Jaroslav Zak
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | - Isaraphorn Pratumchai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Brett S. Marro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | - Kristi L. Marquardt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | | | - Luke L. Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, USA
| | - Michael B. A. Oldstone
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | - Judith A. Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Livia Hegerova
- Division of Hematology, University of Washington School of Medicine, Seattle, USA
| | - Qing Cao
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| | - Umar Farooq
- Division of Hematology and Oncology and Bone Marrow Transplantation, University of Iowa, Iowa City, USA
| | | | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, USA
| | - John R. Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| |
Collapse
|
6
|
Torres-Fernandez D, Dalsuco J, Bramugy J, Bassat Q, Varo R. Innovative strategies for the surveillance, prevention, and management of pediatric infections applied to low-income settings. Expert Rev Anti Infect Ther 2024; 22:413-422. [PMID: 38739471 DOI: 10.1080/14787210.2024.2354839] [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: 06/09/2023] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Infectious diseases still cause a significant burden of morbidity and mortality among children in low- and middle-income countries (LMICs). There are ample opportunities for innovation in surveillance, prevention, and management, with the ultimate goal of improving survival. AREAS COVERED This review discusses the current status in the use and development of innovative strategies for pediatric infectious diseases in LMICs by focusing on surveillance, diagnosis, prevention, and management. Topics covered are: Minimally Invasive Tissue Sampling as a technique to accurately ascertain the cause of death; Genetic Surveillance to trace the pathogen genomic diversity and emergence of resistance; Artificial Intelligence as a multidisciplinary tool; Portable noninvasive imaging methods; and Prognostic Biomarkers to triage and risk stratify pediatric patients. EXPERT OPINION To overcome the specific hurdles in child health for LMICs, some innovative strategies appear at the forefront of research. If the development of these next-generation tools remains focused on accessibility, sustainability and capacity building, reshaping epidemiological surveillance, diagnosis, and treatment in LMICs, can become a reality and result in a significant public health impact. Their integration with existing healthcare infrastructures may revolutionize disease detection and surveillance, and improve child health and survival.
Collapse
Affiliation(s)
- David Torres-Fernandez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Jessica Dalsuco
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Justina Bramugy
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ICREA, Pg. Lluís Companys, Barcelona, Spain
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
- CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosauro Varo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| |
Collapse
|
7
|
Mogire RM, Miruka SA, Juma DW, McNamara CW, Andagalu B, Burrows JN, Chenu E, Duffy J, Ogutu BR, Akala HM. Protein target similarity is positive predictor of in vitro antipathogenic activity: a drug repurposing strategy for Plasmodium falciparum. J Cheminform 2024; 16:63. [PMID: 38831351 PMCID: PMC11145868 DOI: 10.1186/s13321-024-00856-7] [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: 11/26/2023] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
Drug discovery is an intricate and costly process. Repurposing existing drugs and active compounds offers a viable pathway to develop new therapies for various diseases. By leveraging publicly available biomedical information, it is possible to predict compounds' activity and identify their potential targets across diverse organisms. In this study, we aimed to assess the antiplasmodial activity of compounds from the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library using in vitro and bioinformatics approaches. We assessed the in vitro antiplasmodial activity of the compounds using blood-stage and liver-stage drug susceptibility assays. We used protein sequences of known targets of the ReFRAME compounds with high antiplasmodial activity (EC50 < 10 uM) to conduct a protein-pairwise search to identify similar Plasmodium falciparum 3D7 proteins (from PlasmoDB) using NCBI protein BLAST. We further assessed the association between the compounds' in vitro antiplasmodial activity and level of similarity between their known and predicted P. falciparum target proteins using simple linear regression analyses. BLAST analyses revealed 735 P. falciparum proteins that were similar to the 226 known protein targets associated with the ReFRAME compounds. Antiplasmodial activity of the compounds was positively associated with the degree of similarity between the compounds' known targets and predicted P. falciparum protein targets (percentage identity, E value, and bit score), the number of the predicted P. falciparum targets, and their respective mutagenesis index and fitness scores (R2 between 0.066 and 0.92, P < 0.05). Compounds predicted to target essential P. falciparum proteins or those with a druggability index of 1 showed the highest antiplasmodial activity.
Collapse
Affiliation(s)
- Reagan M Mogire
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya.
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya.
| | - Silviane A Miruka
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya
| | - Dennis W Juma
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
- Department of Emerging Infections Diseases (DEID), Walter Reed Army Institute of Research - Africa, Kisumu, Kenya
| | - Case W McNamara
- Calibr-Skaggs Institute for Innovative Medicine, a division of The Scripps Research Institute, La Jolla, CA, USA
| | - Ben Andagalu
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
| | | | - Elodie Chenu
- Medicines for Malaria Venture, Geneva, Switzerland
| | - James Duffy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Bernhards R Ogutu
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya
| | - Hoseah M Akala
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya.
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya.
- Department of Emerging Infections Diseases (DEID), Walter Reed Army Institute of Research - Africa, Kisumu, Kenya.
| |
Collapse
|
8
|
Shao S, Zhang N, Specht GP, You S, Song L, Fu Q, Huang D, You H, Shu J, Domissy A, Li S, Nguyen-Tran V, Joseph SB, Chatterjee AK, Chen JJ, Schultz PG, Bollong MJ. Pharmacological expansion of type 2 alveolar epithelial cells promotes regenerative lower airway repair. Proc Natl Acad Sci U S A 2024; 121:e2400077121. [PMID: 38598345 PMCID: PMC11032444 DOI: 10.1073/pnas.2400077121] [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: 01/03/2024] [Accepted: 03/06/2024] [Indexed: 04/12/2024] Open
Abstract
Type 2 alveolar epithelial cells (AEC2s) are stem cells in the adult lung that contribute to lower airway repair. Agents that promote the selective expansion of these cells might stimulate regeneration of the compromised alveolar epithelium, an etiology-defining event in several pulmonary diseases. From a high-content imaging screen of the drug repurposing library ReFRAME, we identified that dipeptidyl peptidase 4 (DPP4) inhibitors, widely used type 2 diabetes medications, selectively expand AEC2s and are broadly efficacious in several mouse models of lung damage. Mechanism of action studies revealed that the protease DPP4, in addition to processing incretin hormones, degrades IGF-1 and IL-6, essential regulators of AEC2 expansion whose levels are increased in the luminal compartment of the lung in response to drug treatment. To selectively target DPP4 in the lung with sufficient drug exposure, we developed NZ-97, a locally delivered, lung persistent DPP4 inhibitor that broadly promotes efficacy in mouse lung damage models with minimal peripheral exposure and good tolerability. This work reveals DPP4 as a central regulator of AEC2 expansion and affords a promising therapeutic approach to broadly stimulate regenerative repair in pulmonary disease.
Collapse
Affiliation(s)
- Sida Shao
- Calibr, a Division of Scripps Research, La Jolla, CA92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Nan Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Gregory P. Specht
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Shaochen You
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Lirui Song
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | - Qiangwei Fu
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | - David Huang
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | - Hengyao You
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Jian Shu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA02129
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Alain Domissy
- DNA Array Core, The Scripps Research Institute, La Jolla, CA92037
| | - Shuangwei Li
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | | | - Sean B. Joseph
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | | | | | - Peter G. Schultz
- Calibr, a Division of Scripps Research, La Jolla, CA92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| |
Collapse
|
9
|
Hasan MM, Mattice EB, Teixeira JE, Jumani RS, Stebbins EE, Klopfer CE, Franco SE, Love MS, McNamara CW, Huston CD. Cryptosporidium life cycle small molecule probing implicates translational repression and an Apetala 2 transcription factor in macrogamont differentiation. PLoS Pathog 2024; 20:e1011906. [PMID: 38669269 PMCID: PMC11078545 DOI: 10.1371/journal.ppat.1011906] [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: 12/15/2023] [Revised: 05/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The apicomplexan parasite Cryptosporidium is a leading cause of childhood diarrhea in developing countries. Current treatment options are inadequate and multiple preclinical compounds are being actively pursued as potential drugs for cryptosporidiosis. Unlike most apicomplexans, Cryptosporidium spp. sequentially replicate asexually and then sexually within a single host to complete their lifecycles. Anti-cryptosporidial compounds are generally identified or tested through in vitro phenotypic assays that only assess the asexual stages. Therefore, compounds that specifically target the sexual stages remain unexplored. In this study, we leveraged the ReFRAME drug repurposing library against a newly devised multi-readout imaging assay to identify small-molecule compounds that modulate macrogamont differentiation and maturation. RNA-seq studies confirmed selective modulation of macrogamont differentiation for 10 identified compounds (9 inhibitors and 1 accelerator). The collective transcriptomic profiles of these compounds indicates that translational repression accompanies Cryptosporidium sexual differentiation, which we validated experimentally. Additionally, cross comparison of the RNA-seq data with promoter sequence analysis for stage-specific genes converged on a key role for an Apetala 2 (AP2) transcription factor (cgd2_3490) in differentiation into macrogamonts. Finally, drug annotation for the ReFRAME hits indicates that an elevated supply of energy equivalence in the host cell is critical for macrogamont formation.
Collapse
Affiliation(s)
- Muhammad M. Hasan
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| | - Ethan B. Mattice
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| | - José E. Teixeira
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Rajiv S. Jumani
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| | - Erin E. Stebbins
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Connor E. Klopfer
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Sebastian E. Franco
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Melissa S. Love
- Calibr at Scripps Research, San Diego, California, United States of America
| | - Case W. McNamara
- Calibr at Scripps Research, San Diego, California, United States of America
| | - Christopher D. Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| |
Collapse
|
10
|
Maher SP, Bakowski MA, Vantaux A, Flannery EL, Andolina C, Gupta M, Antonova-Koch Y, Argomaniz M, Cabrera-Mora M, Campo B, Chao AT, Chatterjee AK, Cheng WT, Chuenchob E, Cooper CA, Cottier K, Galinski MR, Harupa-Chung A, Ji H, Joseph SB, Lenz T, Lonardi S, Matheson J, Mikolajczak SA, Moeller T, Orban A, Padín-Irizarry V, Pan K, Péneau J, Prudhomme J, Roesch C, Ruberto AA, Sabnis SS, Saney CL, Sattabongkot J, Sereshki S, Suriyakan S, Ubalee R, Wang Y, Wasisakun P, Yin J, Popovici J, McNamara CW, Joyner CJ, Nosten F, Witkowski B, Le Roch KG, Kyle DE. A Drug Repurposing Approach Reveals Targetable Epigenetic Pathways in Plasmodium vivax Hypnozoites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.31.526483. [PMID: 36778461 PMCID: PMC9915689 DOI: 10.1101/2023.01.31.526483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Radical cure of Plasmodium vivax malaria must include elimination of quiescent 'hypnozoite' forms in the liver; however, the only FDA-approved treatments are contraindicated in many vulnerable populations. To identify new drugs and drug targets for hypnozoites, we screened the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library and a collection of epigenetic inhibitors against P. vivax liver stages. From both libraries, we identified inhibitors targeting epigenetics pathways as selectively active against P. vivax and P. cynomolgi hypnozoites. These include DNA methyltransferase (DNMT) inhibitors as well as several inhibitors targeting histone post-translational modifications. Immunofluorescence staining of Plasmodium liver forms showed strong nuclear 5-methylcystosine signal, indicating liver stage parasite DNA is methylated. Using bisulfite sequencing, we mapped genomic DNA methylation in sporozoites, revealing DNA methylation signals in most coding genes. We also demonstrated that methylation level in proximal promoter regions as well as in the first exon of the genes may affect, at least partially, gene expression in P. vivax. The importance of selective inhibitors targeting epigenetic features on hypnozoites was validated using MMV019721, an acetyl-CoA synthetase inhibitor that affects histone acetylation and was previously reported as active against P. falciparum blood stages. In summary, our data indicate that several epigenetic mechanisms are likely modulating hypnozoite formation or persistence and provide an avenue for the discovery and development of improved radical cure antimalarials.
Collapse
Affiliation(s)
- S. P. Maher
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | - M. A. Bakowski
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - A. Vantaux
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - E. L. Flannery
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - C. Andolina
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - M. Gupta
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - Y. Antonova-Koch
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - M. Argomaniz
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - M. Cabrera-Mora
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - B. Campo
- Medicines for Malaria Venture (MMV); Geneva, 1215, Switzerland
| | - A. T. Chao
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - A. K. Chatterjee
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - W. T. Cheng
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - E. Chuenchob
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - C. A. Cooper
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | | | - M. R. Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
- Division of Infectious Diseases, Department of Medicine, Emory University; Atlanta, GA, 30329, USA
| | - A. Harupa-Chung
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - H. Ji
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - S. B. Joseph
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - T. Lenz
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - S. Lonardi
- Department of Computer Science and Engineering, University of California; Riverside, CA, 92521, USA
| | - J. Matheson
- Department of Microbiology and Immunology, University of Otago; Dunedin, 9016, New Zealand
| | - S. A. Mikolajczak
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | | | - A. Orban
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - V. Padín-Irizarry
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
- School of Sciences, Clayton State University; Morrow, GA, 30260, USA
| | - K. Pan
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - J. Péneau
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - J. Prudhomme
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - C. Roesch
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - A. A. Ruberto
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | - S. S. Sabnis
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - C. L. Saney
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - J. Sattabongkot
- Mahidol Vivax Research Unit, Mahidol University; Bangkok, 10400, Thailand
| | - S. Sereshki
- Department of Computer Science and Engineering, University of California; Riverside, CA, 92521, USA
| | - S. Suriyakan
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - R. Ubalee
- Department of Entomology, Armed Forces Research Institute of Medical Sciences (AFRIMS); Bangkok, 10400, Thailand
| | - Y. Wang
- Department of Chemistry, University of California; Riverside, CA, 92521
- Environmental Toxicology Graduate Program, University of California; Riverside, CA, 92521, USA
| | - P. Wasisakun
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - J. Yin
- Environmental Toxicology Graduate Program, University of California; Riverside, CA, 92521, USA
| | - J. Popovici
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - C. W. McNamara
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - C. J. Joyner
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - F. Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford; Oxford, OX3 7LG, UK
| | - B. Witkowski
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - K. G. Le Roch
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - D. E. Kyle
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| |
Collapse
|
11
|
Haid S, Matthaei A, Winkler M, Sake SM, Gunesch AP, Milke V, Köhler NM, Rückert J, Vieyres G, Kühl D, Nguyen TT, Göhl M, Lasswitz L, Zapatero-Belinchón FJ, Brogden G, Gerold G, Wiegmann B, Bilitewski U, Brown RJP, Brönstrup M, Schulz TF, Pietschmann T. Repurposing screen identifies novel candidates for broad-spectrum coronavirus antivirals and druggable host targets. Antimicrob Agents Chemother 2024; 68:e0121023. [PMID: 38319076 PMCID: PMC10916382 DOI: 10.1128/aac.01210-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Libraries composed of licensed drugs represent a vast repertoire of molecules modulating physiological processes in humans, providing unique opportunities for the discovery of host-targeting antivirals. We screened the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) repurposing library with approximately 12,000 molecules for broad-spectrum coronavirus antivirals and discovered 134 compounds inhibiting an alphacoronavirus and mapping to 58 molecular target categories. Dominant targets included the 5-hydroxytryptamine receptor, the dopamine receptor, and cyclin-dependent kinases. Gene knock-out of the drugs' host targets including cathepsin B and L (CTSB/L; VBY-825), the aryl hydrocarbon receptor (AHR; Phortress), the farnesyl-diphosphate farnesyltransferase 1 (FDFT1; P-3622), and the kelch-like ECH-associated protein 1 (KEAP1; Omaveloxolone), significantly modulated HCoV-229E infection, providing evidence that these compounds inhibited the virus through acting on their respective host targets. Counter-screening of all 134 primary compound candidates with SARS-CoV-2 and validation in primary cells identified Phortress, an AHR activating ligand, P-3622-targeting FDFT1, and Omaveloxolone, which activates the NFE2-like bZIP transcription factor 2 (NFE2L2) by liberating it from its endogenous inhibitor KEAP1, as antiviral candidates for both an Alpha- and a Betacoronavirus. This study provides an overview of HCoV-229E repurposing candidates and reveals novel potentially druggable viral host dependency factors hijacked by diverse coronaviruses.
Collapse
Affiliation(s)
- Sibylle Haid
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Alina Matthaei
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Melina Winkler
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Svenja M. Sake
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Antonia P. Gunesch
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Vanessa Milke
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Natalie M. Köhler
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Jessica Rückert
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Hannover, Germany
| | - Gabrielle Vieyres
- Junior Research Group “Cell Biology of RNA Viruses”, Leibniz Institute of Experimental Virology, Hamburg, Germany
- Integrative Analysis of Pathogen-Induced Compartments, Leibniz ScienceCampus InterACt, Hamburg, Germany
| | - David Kühl
- Junior Research Group “Cell Biology of RNA Viruses”, Leibniz Institute of Experimental Virology, Hamburg, Germany
| | - Tu-Trinh Nguyen
- Calibr, a Division of The Scripps Research Institute, La Jolla, California, USA
| | - Matthias Göhl
- German Center for Infection Research, Hannover-Braunschweig Site, Hannover, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lisa Lasswitz
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Francisco J. Zapatero-Belinchón
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Graham Brogden
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gisa Gerold
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Clinical Microbiology, Virology, 901 87 Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), 901 87 Umeå University, Umeå, Sweden
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Bettina Wiegmann
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Hannover Medical School, Hannover, Germany
- BREATH (Biomedical Research in Endstage and Obstructive Lung Disease Hannover), German Center for Lung Research (DZL), Carl-Neuberg Str. 1, Hannover, Germany
| | | | - Richard J. P. Brown
- Division of Veterinary Medicine, Paul Ehrlich Institute, Langen, Germany
- Department of Molecular and Medical Virology, Ruhr University, Bochum, Germany
| | - Mark Brönstrup
- German Center for Infection Research, Hannover-Braunschweig Site, Hannover, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas F. Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Thomas Pietschmann
- Institute for Experimental Virology, Twincore - Centre for Experimental and Clinical Infection Research, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Dayan Elshan NGR, Wolff KC, Riva L, Woods AK, Grabovyi G, Wilson K, Pedroarena J, Ghorai S, Nazarian A, Weiss F, Liu Y, Mazumdar W, Song L, Okwor N, Malvin J, Bakowski MA, Beutler N, Kirkpatrick MG, Gebara-Lamb A, Huang E, Nguyen-Tran VTB, Chi V, Li S, Rogers TF, McNamara CW, Gupta AK, Rahimi A, Chen JJ, Joseph SB, Schultz PG, Chatterjee AK. Discovery of CMX990: A Potent SARS-CoV-2 3CL Protease Inhibitor Bearing a Novel Warhead. J Med Chem 2024; 67:2369-2378. [PMID: 38335279 PMCID: PMC10895651 DOI: 10.1021/acs.jmedchem.3c01938] [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: 10/17/2023] [Revised: 11/26/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
There remains a need to develop novel SARS-CoV-2 therapeutic options that improve upon existing therapies by an increased robustness of response, fewer safety liabilities, and global-ready accessibility. Functionally critical viral main protease (Mpro, 3CLpro) of SARS-CoV-2 is an attractive target due to its homology within the coronaviral family, and lack thereof toward human proteases. In this disclosure, we outline the advent of a novel SARS-CoV-2 3CLpro inhibitor, CMX990, bearing an unprecedented trifluoromethoxymethyl ketone warhead. Compared with the marketed drug nirmatrelvir (combination with ritonavir = Paxlovid), CMX990 has distinctly differentiated potency (∼5× more potent in primary cells) and human in vitro clearance (>4× better microsomal clearance and >10× better hepatocyte clearance), with good in vitro-to-in vivo correlation. Based on its compelling preclinical profile and projected once or twice a day dosing supporting unboosted oral therapy in humans, CMX990 advanced to a Phase 1 clinical trial as an oral drug candidate for SARS-CoV-2.
Collapse
Affiliation(s)
- N. G. R. Dayan Elshan
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Karen C. Wolff
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Laura Riva
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Ashley K. Woods
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Gennadii Grabovyi
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Katy Wilson
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - James Pedroarena
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Sourav Ghorai
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Armen Nazarian
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Frank Weiss
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Yuyin Liu
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Wrickban Mazumdar
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lirui Song
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Neechi Okwor
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Jacqueline Malvin
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Malina A. Bakowski
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Nathan Beutler
- Department
of Immunology and Microbiology, The Scripps
Research Institute, 10466
North Torrey Pines Road, La Jolla, California 92037, United States
| | - Melanie G. Kirkpatrick
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Amal Gebara-Lamb
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Edward Huang
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Vân T. B. Nguyen-Tran
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Victor Chi
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Shuangwei Li
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Thomas F. Rogers
- Department
of Immunology and Microbiology, The Scripps
Research Institute, 10466
North Torrey Pines Road, La Jolla, California 92037, United States
| | - Case W. McNamara
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Anil Kumar Gupta
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Alireza Rahimi
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Jian Jeffrey Chen
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Sean B. Joseph
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Peter G. Schultz
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
- Department
of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Arnab K. Chatterjee
- Calibr
at Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
13
|
Stanton C, Sun J, Nutsch K, Rosarda JD, Nguyen T, Li-Ma C, Njomen E, Kutseikin S, Saez E, Teijaro JR, Wiseman RL, Bollong MJ. Covalent Targeting As a Common Mechanism for Inhibiting NLRP3 Inflammasome Assembly. ACS Chem Biol 2024; 19:254-265. [PMID: 38198472 PMCID: PMC11131128 DOI: 10.1021/acschembio.3c00330] [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] [Indexed: 01/12/2024]
Abstract
The NLRP3 inflammasome is a cytosolic protein complex important for the regulation and secretion of inflammatory cytokines, including IL-1β and IL-18. Aberrant overactivation of NLRP3 is implicated in numerous inflammatory disorders. However, the activation and regulation of NLRP3 inflammasome signaling remain poorly understood, limiting our ability to develop pharmacologic approaches to target this important inflammatory complex. Here, we developed and implemented a high-throughput screen to identify compounds that inhibit the inflammasome assembly and activity. From this screen, we identify and profile inflammasome inhibition of 20 new covalent compounds across nine different chemical scaffolds, as well as many known inflammasome covalent inhibitors. Intriguingly, our results indicate that NLRP3 possesses numerous reactive cysteines on multiple domains whose covalent targeting blocks the activation of this inflammatory complex. Specifically, focusing on compound VLX1570, which possesses multiple electrophilic moieties, we demonstrate that this compound allows covalent, intermolecular cross-linking of NLRP3 cysteines to inhibit inflammasome assembly. Our results, along with the recent identification of numerous covalent molecules that inhibit NLRP3 inflammasome activation, further support the continued development of electrophilic compounds that target reactive cysteine residues on NLRP3 to regulate its activation and activity.
Collapse
Affiliation(s)
- Caroline Stanton
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jie Sun
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jessica D. Rosarda
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Thu Nguyen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Chloris Li-Ma
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Evert Njomen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Sergei Kutseikin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - John R. Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
14
|
Sake SM, Zhang X, Rajak MK, Urbanek-Quaing M, Carpentier A, Gunesch AP, Grethe C, Matthaei A, Rückert J, Galloux M, Larcher T, Le Goffic R, Hontonnou F, Chatterjee AK, Johnson K, Morwood K, Rox K, Elgaher WAM, Huang J, Wetzke M, Hansen G, Fischer N, Eléouët JF, Rameix-Welti MA, Hirsch AKH, Herold E, Empting M, Lauber C, Schulz TF, Krey T, Haid S, Pietschmann T. Drug repurposing screen identifies lonafarnib as respiratory syncytial virus fusion protein inhibitor. Nat Commun 2024; 15:1173. [PMID: 38332002 PMCID: PMC10853176 DOI: 10.1038/s41467-024-45241-y] [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: 12/19/2022] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of acute lower respiratory tract infection in infants, older adults and the immunocompromised. Effective directly acting antivirals are not yet available for clinical use. To address this, we screen the ReFRAME drug-repurposing library consisting of 12,000 small molecules against RSV. We identify 21 primary candidates including RSV F and N protein inhibitors, five HSP90 and four IMPDH inhibitors. We select lonafarnib, a licensed farnesyltransferase inhibitor, and phase III candidate for hepatitis delta virus (HDV) therapy, for further follow-up. Dose-response analyses and plaque assays confirm the antiviral activity (IC50: 10-118 nM). Passaging of RSV with lonafarnib selects for phenotypic resistance and fixation of mutations in the RSV fusion protein (T335I and T400A). Lentiviral pseudotypes programmed with variant RSV fusion proteins confirm that lonafarnib inhibits RSV cell entry and that these mutations confer lonafarnib resistance. Surface plasmon resonance reveals RSV fusion protein binding of lonafarnib and co-crystallography identifies the lonafarnib binding site within RSV F. Oral administration of lonafarnib dose-dependently reduces RSV virus load in a murine infection model using female mice. Collectively, this work provides an overview of RSV drug repurposing candidates and establishes lonafarnib as a bona fide fusion protein inhibitor.
Collapse
Affiliation(s)
- Svenja M Sake
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Xiaoyu Zhang
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Manoj Kumar Rajak
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck, Luebeck, Germany
| | - Melanie Urbanek-Quaing
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Arnaud Carpentier
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Antonia P Gunesch
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Christina Grethe
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Alina Matthaei
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Jessica Rückert
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Marie Galloux
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | | | - Ronan Le Goffic
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | | | | | | | | | - Katharina Rox
- Department of Chemical Biology, Helmholtz Center of Infection Research, Braunschweig, Germany
- German Centre for Infection Research, Partner site Braunschweig-Hannover, Braunschweig, Germany
| | - Walid A M Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-HZI, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Jiabin Huang
- Insitute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Wetzke
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Partner Site Hannover, BREATH, Hannover, Germany
| | - Gesine Hansen
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Partner Site Hannover, BREATH, Hannover, Germany
| | - Nicole Fischer
- Insitute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Marie-Anne Rameix-Welti
- Université Paris-Saclay, Université de Versailles St. Quentin; UMR 1173 (2I), INSERM; Assistance Publique des Hôpitaux de Paris, Hôpital Ambroise Paré, Laboratoire de Microbiologie, DMU15, Versailles, France
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-HZI, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Helmholtz International Lab for Anti-infectives, HZI, Braunschweig, Germany
| | - Elisabeth Herold
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck, Luebeck, Germany
| | - Martin Empting
- German Centre for Infection Research, Partner site Braunschweig-Hannover, Braunschweig, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-HZI, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Chris Lauber
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research, Partner site Braunschweig-Hannover, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Thomas Krey
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck, Luebeck, Germany
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Luebeck-Borstel-Riems, Luebeck, Germany
| | - Sibylle Haid
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.
| | - Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany.
- German Centre for Infection Research, Partner site Braunschweig-Hannover, Braunschweig, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
- Helmholtz International Lab for Anti-infectives, HZI, Braunschweig, Germany.
| |
Collapse
|
15
|
Abstract
The concept of drug repurposing is focused on the repositioning of drug molecules that have already undergone safety trials. There are different strategies for drug repurposing. Network-based strategy focuses on the evaluation of drug combinations in a molecular environment with multi-target hits and analysis of drug interactions. Implementation of any in silico strategy requires several databases and pipelines for executing the process of shortlisting appropriate drugs.
Collapse
Affiliation(s)
- Arjun V Kowshik
- Department of Biotechnology, PES University, Bengaluru, India
| | - Megha Manoj
- Department of Biotechnology, PES University, Bengaluru, India
| | | | | |
Collapse
|
16
|
Clare RH, Dawson CA, Westhorpe A, Albulescu LO, Woodley CM, Mosallam N, Chong DJW, Kool J, Berry NG, O’Neill PM, Casewell NR. Snakebite drug discovery: high-throughput screening to identify novel snake venom metalloproteinase toxin inhibitors. Front Pharmacol 2024; 14:1328950. [PMID: 38273820 PMCID: PMC10808794 DOI: 10.3389/fphar.2023.1328950] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Snakebite envenoming results in ∼100,000 deaths per year, with close to four times as many victims left with life-long sequelae. Current antivenom therapies have several limitations including high cost, variable cross-snake species efficacy and a requirement for intravenous administration in a clinical setting. Next-generation snakebite therapies are being widely investigated with the aim to improve cost, efficacy, and safety. In recent years several small molecule drugs have shown considerable promise for snakebite indication, with oral bioavailability particularly promising for community delivery rapidly after a snakebite. However, only two such drugs have entered clinical development for snakebite. To offset the risk of attrition during clinical trials and to better explore the chemical space for small molecule venom toxin inhibitors, here we describe the first high throughput drug screen against snake venom metalloproteinases (SVMPs)-a pathogenic toxin family responsible for causing haemorrhage and coagulopathy. Following validation of a 384-well fluorescent enzymatic assay, we screened a repurposed drug library of 3,547 compounds against five geographically distinct and toxin variable snake venoms. Our drug screen resulted in the identification of 14 compounds with pan-species inhibitory activity. Following secondary potency testing, four SVMP inhibitors were identified with nanomolar EC50s comparable to the previously identified matrix metalloproteinase inhibitor marimastat and superior to the metal chelator dimercaprol, doubling the current global portfolio of SVMP inhibitors. Following analysis of their chemical structure and ADME properties, two hit-to-lead compounds were identified. These clear starting points for the initiation of medicinal chemistry campaigns provide the basis for the first ever designer snakebite specific small molecules.
Collapse
Affiliation(s)
- Rachel H. Clare
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Charlotte A. Dawson
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Adam Westhorpe
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Laura-Oana Albulescu
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Nada Mosallam
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Daniel J. W. Chong
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Neil G. Berry
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Paul M. O’Neill
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Nicholas R. Casewell
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
17
|
Olmedo DA, Durant-Archibold AA, López-Pérez JL, Medina-Franco JL. Design and Diversity Analysis of Chemical Libraries in Drug Discovery. Comb Chem High Throughput Screen 2024; 27:502-515. [PMID: 37409545 DOI: 10.2174/1386207326666230705150110] [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: 04/05/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023]
Abstract
Chemical libraries and compound data sets are among the main inputs to start the drug discovery process at universities, research institutes, and the pharmaceutical industry. The approach used in the design of compound libraries, the chemical information they possess, and the representation of structures, play a fundamental role in the development of studies: chemoinformatics, food informatics, in silico pharmacokinetics, computational toxicology, bioinformatics, and molecular modeling to generate computational hits that will continue the optimization process of drug candidates. The prospects for growth in drug discovery and development processes in chemical, biotechnological, and pharmaceutical companies began a few years ago by integrating computational tools with artificial intelligence methodologies. It is anticipated that it will increase the number of drugs approved by regulatory agencies shortly.
Collapse
Affiliation(s)
- Dionisio A Olmedo
- Centro de Investigaciones Farmacognósticas de la Flora Panameña (CIFLORPAN), Facultad de Farmacia, Universidad de Panamá, Ciudad de Panamá, Apartado, 0824-00178, Panamá
- Sistema Nacional de Investigación (SNI), Secretaria Nacional de Ciencia, Tecnología e Innovación (SENACYT), Ciudad del Saber, Clayton, Panamá
| | - Armando A Durant-Archibold
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Apartado, 0843-01103, Panamá
- Departamento de Bioquímica, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Ciudad de Panamá, Panamá
| | - José Luis López-Pérez
- CESIFAR, Departamento de Farmacología, Facultad de Medicina, Universidad de Panamá, Ciudad de Panamá, Panamá
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, Avda. Campo Charro s/n, 37071 Salamanca, España
| | - José Luis Medina-Franco
- DIFACQUIM Grupo de Investigación, Departamento de Farmacia, Escuela de Química, Universidad Nacional Autónoma de México, Ciudad de México, Apartado, 04510, México
| |
Collapse
|
18
|
Hakmi M, Bouricha EM, Soussi A, Bzioui IA, Belyamani L, Ibrahimi A. Computational Drug Design Strategies for Fighting the COVID-19 Pandemic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1457:199-214. [PMID: 39283428 DOI: 10.1007/978-3-031-61939-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The advent of COVID-19 has brought the use of computer tools to the fore in health research. In recent years, computational methods have proven to be highly effective in a variety of areas, including genomic surveillance, host range prediction, drug target identification, and vaccine development. They were also instrumental in identifying new antiviral compounds and repurposing existing therapeutics to treat COVID-19. Using computational approaches, researchers have made significant advances in understanding the molecular mechanisms of COVID-19 and have developed several promising drug candidates and vaccines. This chapter highlights the critical importance of computational drug design strategies in elucidating various aspects of COVID-19 and their contribution to advancing global drug design efforts during the pandemic. Ultimately, the use of computing tools will continue to play an essential role in health research, enabling researchers to develop innovative solutions to combat new and emerging diseases.
Collapse
Affiliation(s)
- Mohammed Hakmi
- Medical Biotechnology Laboratory (MedBiotech), Faculty of Medicine and Pharmacy, Bioinova Research Center, Mohammed Vth University, Rabat, Morocco.
- Mohammed VI Center for Research and Innovation (CM6), Rabat, Morocco.
| | - El Mehdi Bouricha
- Medical Biotechnology Laboratory (MedBiotech), Faculty of Medicine and Pharmacy, Bioinova Research Center, Mohammed Vth University, Rabat, Morocco
- Mohammed VI Center for Research and Innovation (CM6), Rabat, Morocco
| | - Abdellatif Soussi
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16145, Genova, Italy
| | - Ilias Abdeslam Bzioui
- Department of Gynecology and Obstetrics, Faculty of Medicine, Abdelmalek Essaâdi University Hospital, Tangier, Morocco
| | - Lahcen Belyamani
- Mohammed VI Center for Research and Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
- Emergency Department, Medical and Pharmacy School, Military Hospital Mohammed V, Mohammed V University, Rabat, Morocco
| | - Azeddine Ibrahimi
- Medical Biotechnology Laboratory (MedBiotech), Faculty of Medicine and Pharmacy, Bioinova Research Center, Mohammed Vth University, Rabat, Morocco
- Mohammed VI Center for Research and Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| |
Collapse
|
19
|
Bulos ML, Grzelak EM, Li-Ma C, Chen E, Hull M, Johnson KA, Bollong MJ. Pharmacological inhibition of CLK2 activates YAP by promoting alternative splicing of AMOTL2. eLife 2023; 12:RP88508. [PMID: 38126343 PMCID: PMC10735217 DOI: 10.7554/elife.88508] [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] [Indexed: 12/23/2023] Open
Abstract
Yes-associated protein (YAP), the downstream effector of the evolutionarily conserved Hippo pathway, promotes cellular proliferation and coordinates certain regenerative responses in mammals. Small molecule activators of YAP may, therefore, display therapeutic utility in treating disease states involving insufficient proliferative repair. From a high-throughput chemical screen of the comprehensive drug repurposing library ReFRAME, here we report the identification of SM04690, a clinical stage inhibitor of CLK2, as a potent activator of YAP-driven transcriptional activity in cells. CLK2 inhibition promotes alternative splicing of the Hippo pathway protein AMOTL2, producing an exon-skipped gene product that can no longer associate with membrane-bound proteins, resulting in decreased phosphorylation and membrane localization of YAP. This study reveals a novel mechanism by which pharmacological perturbation of alternative splicing inactivates the Hippo pathway and promotes YAP-dependent cellular growth.
Collapse
Affiliation(s)
- Maya L Bulos
- Department of Chemistry, The Scripps Research InstituteLa JollaUnited States
| | - Edyta M Grzelak
- Department of Chemistry, The Scripps Research InstituteLa JollaUnited States
| | - Chloris Li-Ma
- Department of Chemistry, The Scripps Research InstituteLa JollaUnited States
| | - Emily Chen
- Calibr, A Division of Scripps ResearchLa JollaUnited States
| | - Mitchell Hull
- Calibr, A Division of Scripps ResearchLa JollaUnited States
| | | | - Michael J Bollong
- Department of Chemistry, The Scripps Research InstituteLa JollaUnited States
| |
Collapse
|
20
|
Meyer SM, Tanaka T, Taghavi A, Baisden JT, Grefe M, Disney MD. Optimization of a Protein-Targeted Medicine into an RNA-Specific Small Molecule. ACS Chem Biol 2023; 18:2336-2342. [PMID: 37870980 PMCID: PMC10825933 DOI: 10.1021/acschembio.3c00476] [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] [Indexed: 10/25/2023]
Abstract
Protein-targeted small molecule medicines often bind RNAs and affect RNA-mediated pathways in cells. Historically, small molecule engagement and modulation of RNA have not been considered in medicine development; however, RNA should be considered both a potential on- and off-target. Kinase inhibitors have emecrged as common RNA binders with dovitinib, a classic receptor tyrosine kinase (RTK) inhibitor, inhibiting RTKs and the biogenesis of oncogenic microRNA-21 through direct engagement. In this study, we use knowledge of the molecular recognition of both protein and RNA targets by dovitinib to design molecules that specifically inhibit the RNA target but lack activity against canonical protein targets in cells. As it is now becoming apparent that RNA can be both an on- and off-target for small molecule medicines, this study lays a foundation to use design principles to maximize desired compound activity while minimizing off-target effects.
Collapse
Affiliation(s)
- Samantha M. Meyer
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology and The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
- The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Toru Tanaka
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology and The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Amirhossein Taghavi
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology and The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
- The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Jared T. Baisden
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology and The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Maison Grefe
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology and The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Matthew D. Disney
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology and The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
- The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, FL 33458 USA
| |
Collapse
|
21
|
Nutsch K, Song L, Chen E, Hull M, Chatterjee AK, Chen JJ, Bollong MJ. A covalent inhibitor of the YAP-TEAD transcriptional complex identified by high-throughput screening. RSC Chem Biol 2023; 4:894-905. [PMID: 37920398 PMCID: PMC10619132 DOI: 10.1039/d3cb00044c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/19/2023] [Indexed: 11/04/2023] Open
Abstract
Yes-associated protein (YAP), the master transcriptional effector downstream of the Hippo pathway, regulates essential cell growth and regenerative processes in animals. However, the activation of YAP observed in cancers drives cellular proliferation, metastasis, chemoresistance, and immune suppression, making it of key interest in developing precision therapeutics for oncology. As such, pharmacological inhibition of YAP by targeting its essential co-regulators, TEA domain transcription factors (TEADs) would likely promote tumor clearance in sensitive tumor types. From a fluorescence polarization-based high throughput screen of over 800 000 diverse small molecules, here we report the identification of a pyrazolopyrimidine-based scaffold that inhibits association of YAP and TEADs. Medicinal chemistry-based optimization identified mCMY020, a potent, covalent inhibitor of TEAD transcriptional activity that occupies a conserved, central palmitoylation site on TEADs.
Collapse
Affiliation(s)
- Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Lirui Song
- Calibr, A Division of Scripps Research La Jolla CA 92037 USA
| | - Emily Chen
- Calibr, A Division of Scripps Research La Jolla CA 92037 USA
| | - Mitchell Hull
- Calibr, A Division of Scripps Research La Jolla CA 92037 USA
| | | | | | - Michael J Bollong
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| |
Collapse
|
22
|
van der Pol KH, Aljofan M, Blin O, Cornel JH, Rongen GA, Woestelandt AG, Spedding M. Drug Repurposing of Generic Drugs: Challenges and the Potential Role for Government. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2023; 21:831-840. [PMID: 37398987 PMCID: PMC10627937 DOI: 10.1007/s40258-023-00816-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/04/2023]
Abstract
Drug repurposing is the process of identifying a new use for an existing drug or active substance in an indication outside the scope of the original indication. Drug repurposing has important advantages including reduced development time and costs, and potentially large societal healthcare cost savings. However, current generic drug repurposing research faces a number of challenges in obtaining research funds. Furthermore, regardless of the success of a repurposing trial, commercial parties often lack interest in pursuing marketing authorisation for financial reasons, and academic researchers lack the knowledge, time and funding. Therefore, the new indication of a repurposed drug often does not make it 'on label'. We propose a large increase in public funding for generic drug repurposing research, including funds for the marketing authorisation process when a trial is successful, and a reduction in the regulatory burden of the marketing authorisation process for repurposed generic drugs.
Collapse
Affiliation(s)
- Karel H van der Pol
- Department of Internal Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mohamad Aljofan
- Department of Biomedical Science, Nazarbayev University School of Medicine, Nur-Sultan, Kazakhstan
- National Laboratory Astana, Nazarbayev University, Astana, 010000, Kazakhstan
| | - Olivier Blin
- Institut de Neurosciences des Systèmes, Aix Marseille Université, Inserm UMR 1106, Marseille, France
| | - Jan H Cornel
- Department of Cardiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Gerard A Rongen
- Department of Internal Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | | | |
Collapse
|
23
|
Bertin P, Rector-Brooks J, Sharma D, Gaudelet T, Anighoro A, Gross T, Martínez-Peña F, Tang EL, Suraj MS, Regep C, Hayter JBR, Korablyov M, Valiante N, van der Sloot A, Tyers M, Roberts CES, Bronstein MM, Lairson LL, Taylor-King JP, Bengio Y. RECOVER identifies synergistic drug combinations in vitro through sequential model optimization. CELL REPORTS METHODS 2023; 3:100599. [PMID: 37797618 PMCID: PMC10626197 DOI: 10.1016/j.crmeth.2023.100599] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 10/07/2023]
Abstract
For large libraries of small molecules, exhaustive combinatorial chemical screens become infeasible to perform when considering a range of disease models, assay conditions, and dose ranges. Deep learning models have achieved state-of-the-art results in silico for the prediction of synergy scores. However, databases of drug combinations are biased toward synergistic agents and results do not generalize out of distribution. During 5 rounds of experimentation, we employ sequential model optimization with a deep learning model to select drug combinations increasingly enriched for synergism and active against a cancer cell line-evaluating only ∼5% of the total search space. Moreover, we find that learned drug embeddings (using structural information) begin to reflect biological mechanisms. In silico benchmarking suggests search queries are ∼5-10× enriched for highly synergistic drug combinations by using sequential rounds of evaluation when compared with random selection or ∼3× when using a pretrained model.
Collapse
Affiliation(s)
- Paul Bertin
- Mila, the Quebec AI Institute, Montreal, QC, Canada
| | | | | | | | | | | | | | - Eileen L Tang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | - Almer van der Sloot
- IRIC, Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
| | - Mike Tyers
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | | | - Michael M Bronstein
- Relation Therapeutics, London, UK; Department of Computer Science, University of Oxford, Oxford, UK
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | | |
Collapse
|
24
|
You S, Bollong MJ. A high throughput screen for pharmacological inhibitors of the carbohydrate response element. Sci Data 2023; 10:676. [PMID: 37794069 PMCID: PMC10550954 DOI: 10.1038/s41597-023-02596-z] [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: 04/04/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
A central regulator of metabolism, transcription factor carbohydrate response element binding protein (ChREBP) senses and responds to dietary glucose levels by stimulating the transcription of glycolytic and lipogenic enzymes. Genetic depletion of ChREBP rescues β-cell dysfunction arising from high glucose levels, suggesting that inhibiting ChREBP might represent an attractive therapeutic approach to manage diabetes and other metabolic diseases. However, the molecular mechanisms governing ChREBP activation are poorly understood and chemical tools to probe the cellular activity of ChREBP are lacking. Here, we report a high-throughput pharmacological screen in INS-1E β-cells that identified novel inhibitors of ChREBP-driven transcription at carbohydrate response element sites, including three putative covalent inhibitors and two likely non-covalent chemical scaffolds. This work affords a pharmacological toolkit to help uncover the signaling logic controlling ChREBP activation and may ultimately reveal potential therapeutic approaches for treating metabolic disease.
Collapse
Affiliation(s)
- Shaochen You
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, 92037, USA
| | - Michael J Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, 92037, USA.
| |
Collapse
|
25
|
Kawata M, McClatchy DB, Diedrich JK, Olmer M, Johnson KA, Yates JR, Lotz MK. Mocetinostat activates Krüppel-like factor 4 and protects against tissue destruction and inflammation in osteoarthritis. JCI Insight 2023; 8:e170513. [PMID: 37681413 PMCID: PMC10544226 DOI: 10.1172/jci.insight.170513] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/19/2023] [Indexed: 09/09/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder, and disease-modifying OA drugs (DMOADs) represent a major need in OA management. Krüppel-like factor 4 (KLF4) is a central transcription factor upregulating regenerative and protective functions in joint tissues. This study was aimed to identify small molecules activating KLF4 expression and to determine functions and mechanisms of the hit compounds. High-throughput screening (HTS) with 11,948 clinical-stage compounds was performed using a reporter cell line detecting endogenous KLF4 activation. Eighteen compounds were identified through the HTS and confirmed in a secondary screen. After testing in SW1353 chondrosarcoma cells and human chondrocytes, mocetinostat - a class I selective histone deacetylase (HDAC) inhibitor - had the best profile of biological activities. Mocetinostat upregulated cartilage signature genes in human chondrocytes, meniscal cells, and BM-derived mesenchymal stem cells, and it downregulated hypertrophic, inflammatory, and catabolic genes in those cells and synoviocytes. I.p. administration of mocetinostat into mice reduced severity of OA-associated changes and improved pain behaviors. Global gene expression and proteomics analyses revealed that regenerative and protective effects of mocetinostat were dependent on peroxisome proliferator-activated receptor γ coactivator 1-α. These findings show therapeutic and protective activities of mocetinostat against OA, qualifying it as a candidate to be used as a DMOAD.
Collapse
Affiliation(s)
- Manabu Kawata
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Daniel B. McClatchy
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Jolene K. Diedrich
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Merissa Olmer
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | | | - John R. Yates
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Martin K. Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
26
|
Bulos ML, Grzelak EM, Li-Ma C, Chen E, Hull M, Johnson KA, Bollong MJ. Pharmacological inhibition of CLK2 activates YAP by promoting alternative splicing of AMOTL2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537449. [PMID: 37131806 PMCID: PMC10153145 DOI: 10.1101/2023.04.19.537449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Yes-associated protein (YAP), the downstream effector of the evolutionarily conserved Hippo pathway, promotes cellular proliferation and coordinates certain regenerative responses in mammals. Small molecule activators of YAP may therefore display therapeutic utility in treating disease states involving insufficient proliferative repair. From a high-throughput chemical screen of the comprehensive drug repurposing library ReFRAME, here we report the identification of SM04690, a clinical stage inhibitor of CLK2, as a potent activator of YAP driven transcriptional activity in cells. CLK2 inhibition promotes alternative splicing of the Hippo pathway protein AMOTL2, producing an exon-skipped gene product that can no longer associate with membrane-bound proteins, resulting in decreased phosphorylation and membrane localization of YAP. This study reveals a novel mechanism by which pharmacological perturbation of alternative splicing inactivates the Hippo pathway and promotes YAP dependent cellular growth.
Collapse
Affiliation(s)
- Maya L. Bulos
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Edyta M. Grzelak
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Chloris Li-Ma
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Emily Chen
- Calibr, A Division of Scripps Research, La Jolla, CA, 92037, USA
| | - Mitchell Hull
- Calibr, A Division of Scripps Research, La Jolla, CA, 92037, USA
| | | | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
27
|
Coghlan A, Partridge FA, Duque-Correa MA, Rinaldi G, Clare S, Seymour L, Brandt C, Mkandawire TT, McCarthy C, Holroyd N, Nick M, Brown AE, Tonitiwong S, Sattelle DB, Berriman M. A drug repurposing screen for whipworms informed by comparative genomics. PLoS Negl Trop Dis 2023; 17:e0011205. [PMID: 37669291 PMCID: PMC10503962 DOI: 10.1371/journal.pntd.0011205] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/15/2023] [Accepted: 07/06/2023] [Indexed: 09/07/2023] Open
Abstract
Hundreds of millions of people worldwide are infected with the whipworm Trichuris trichiura. Novel treatments are urgently needed as current drugs, such as albendazole, have relatively low efficacy. We have investigated whether drugs approved for other human diseases could be repurposed as novel anti-whipworm drugs. In a previous comparative genomics analysis, we identified 409 drugs approved for human use that we predicted to target parasitic worm proteins. Here we tested these ex vivo by assessing motility of adult worms of Trichuris muris, the murine whipworm, an established model for human whipworm research. We identified 14 compounds with EC50 values of ≤50 μM against T. muris ex vivo, and selected nine for testing in vivo. However, the best worm burden reduction seen in mice was just 19%. The high number of ex vivo hits against T. muris shows that we were successful at predicting parasite proteins that could be targeted by approved drugs. In contrast, the low efficacy of these compounds in mice suggest challenges due to their chemical properties (e.g. lipophilicity, polarity, molecular weight) and pharmacokinetics (e.g. absorption, distribution, metabolism, and excretion) that may (i) promote absorption by the host gastrointestinal tract, thereby reducing availability to the worms embedded in the large intestine, and/or (ii) restrict drug uptake by the worms. This indicates that identifying structural analogues that have reduced absorption by the host, and increased uptake by worms, may be necessary for successful drug development against whipworms.
Collapse
Affiliation(s)
- Avril Coghlan
- Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Frederick A. Partridge
- University College London, London, United Kingdom
- School of Life Sciences, University of Westminster, London, United Kingdom
| | | | | | - Simon Clare
- Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Lisa Seymour
- Wellcome Sanger Institute, Cambridge, United Kingdom
| | | | | | | | - Nancy Holroyd
- Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Marina Nick
- University College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
28
|
Chiem K, Nogales A, Lorenzo M, Morales Vasquez D, Xiang Y, Gupta YK, Blasco R, de la Torre JC, Martínez-Sobrido L. Identification of In Vitro Inhibitors of Monkeypox Replication. Microbiol Spectr 2023; 11:e0474522. [PMID: 37278625 PMCID: PMC10434227 DOI: 10.1128/spectrum.04745-22] [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: 11/19/2022] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
Monkeypox virus (MPXV) infections in humans have historically been restricted to regions of endemicity in Africa. However, in 2022, an alarming number of MPXV cases were reported globally, with evidence of person-to-person transmission. Because of this, the World Health Organization (WHO) declared the MPXV outbreak a public health emergency of international concern. The supply of MPXV vaccines is limited, and only two antivirals, tecovirimat and brincidofovir, approved by the U.S. Food and Drug Administration (FDA) for the treatment of smallpox, are currently available for the treatment of MPXV infection. Here, we evaluated 19 compounds previously shown to inhibit different RNA viruses for their ability to inhibit orthopoxvirus infections. We first used recombinant vaccinia virus (rVACV) expressing fluorescence (mScarlet or green fluorescent protein [GFP]) and luciferase (Nluc) reporter genes to identify compounds with antiorthopoxvirus activity. Seven compounds from the ReFRAME library (antimycin A, mycophenolic acid, AVN-944, pyrazofurin, mycophenolate mofetil, azaribine, and brequinar) and six compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) showed inhibitory activity against rVACV. Notably, the anti-VACV activity of some of the compounds in the ReFRAME library (antimycin A, mycophenolic acid, AVN-944, mycophenolate mofetil, and brequinar) and all the compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) were confirmed with MPXV, demonstrating their inhibitory activity in vitro against two orthopoxviruses. IMPORTANCE Despite the eradication of smallpox, some orthopoxviruses remain important human pathogens, as exemplified by the recent 2022 monkeypox virus (MPXV) outbreak. Although smallpox vaccines are effective against MPXV, access to those vaccines is limited. In addition, current antiviral treatment against MPXV infections is limited to the use of the FDA-approved drugs tecovirimat and brincidofovir. Thus, there is an urgent need to identify novel antivirals for the treatment of MPXV infection and other potentially zoonotic orthopoxvirus infections. Here, we show that 13 compounds, derived from two different libraries, previously found to inhibit several RNA viruses, also inhibit VACV. Notably, 11 compounds also displayed inhibitory activity against MPXV.
Collapse
Affiliation(s)
- Kevin Chiem
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Aitor Nogales
- Animal Health Research Centre, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Maria Lorenzo
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | | | - Yan Xiang
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yogesh K. Gupta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rafael Blasco
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
29
|
Roquini V, Mengarda AC, Cajas RA, Martins-da-Silva MF, Godoy-Silva J, Santos GA, Espírito-Santo MCC, Pavani TFA, Melo VA, Salvadori MC, Teixeira FS, Rando DGG, de Moraes J. The Existing Drug Nifuroxazide as an Antischistosomal Agent: In Vitro, In Vivo, and In Silico Studies of Macromolecular Targets. Microbiol Spectr 2023; 11:e0139323. [PMID: 37409934 PMCID: PMC10434008 DOI: 10.1128/spectrum.01393-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023] Open
Abstract
Schistosomiasis is a parasitic disease that afflicts approximately 250 million people worldwide. There is an urgent demand for new antiparasitic agents because praziquantel, the only drug available for the treatment of schistosomiasis, is not universally effective and may derail current progress toward the WHO goal of eliminating this disease as a public health problem by 2030. Nifuroxazide (NFZ), an oral nitrofuran antibiotic, has recently been explored to be repurposed for parasitic diseases. Here, in vitro, in vivo, and in silico studies were conducted to evaluate the activity of NFZ on Schistosoma mansoni. The in vitro study showed significant antiparasitic activity, with 50% effective concentration (EC50) and 90% effective concentration (EC90) values of 8.2 to 10.8 and 13.7 to 19.3 μM, respectively. NFZ also affected worm pairing and egg production and induced severe damage to the tegument of schistosomes. In vivo, a single oral dose of NFZ (400 mg/kg of body weight) to mice harboring either prepatent or patent S. mansoni infection significantly reduced the total worm burden (~40%). In patent infection, NFZ achieved a high reduction in the number of eggs (~80%), but the drug caused a low reduction in the egg burden of animals with prepatent infection. Finally, results from in silico target fishing methods predicted that serine/threonine kinases could be one of the potential targets for NFZ in S. mansoni. Overall, the present study revealed that NFZ possesses antischistosomal properties, mainly in terms of egg burden reduction in animals with patent S. mansoni infection. IMPORTANCE The increasing recognition of the burden imposed by helminthiasis, associated with the limited therapeutic arsenal, has led to initiatives and strategies to research and develop new drugs for the treatment of schistosomiasis. One of these strategies is drug repurposing, which considers low-risk compounds with potentially reduced costs and shorter time for development. In this study, nifuroxazide (NFZ) was evaluated for its anti-Schistosoma mansoni potential through in vitro, in vivo, and in silico studies. In vitro, NFZ affected worm pairing and egg production and induced severe damage to the tegument of schistosomes. In vivo, a single oral dose of NFZ (400 mg/kg) to mice harboring either prepatent or patent S. mansoni infection significantly reduced the total worm burden and egg production. In silico investigations have identified serine/threonine kinases as a molecular target for NFZ. Collectively, these results implied that NFZ might be a potential therapeutic candidate for the treatment of schistosomiasis.
Collapse
Affiliation(s)
- Vinícius Roquini
- Research Center on Neglected Diseases, Guarulhos University, Guarulhos, São Paulo, Brazil
| | - Ana C. Mengarda
- Research Center on Neglected Diseases, Guarulhos University, Guarulhos, São Paulo, Brazil
| | - Rayssa A. Cajas
- Research Center on Neglected Diseases, Guarulhos University, Guarulhos, São Paulo, Brazil
| | | | - Julia Godoy-Silva
- Research Center on Neglected Diseases, Guarulhos University, Guarulhos, São Paulo, Brazil
| | - Gustavo A. Santos
- Research Center on Neglected Diseases, Guarulhos University, Guarulhos, São Paulo, Brazil
| | - Maria Cristina C. Espírito-Santo
- Laboratory of Immunopathology of Schistosomiasis (LIM-06), Department of Infectious and Parasitic Diseases, Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
- Laboratory of Helminthology, Institute of Tropical Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Thais F. A. Pavani
- Biological Chemistry Post-Graduate Course, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Vanusa A. Melo
- Biological Chemistry Post-Graduate Course, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Maria C. Salvadori
- Institute of Physics, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Daniela G. G. Rando
- Chemico-Pharmaceutical Research Group, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Josué de Moraes
- Research Center on Neglected Diseases, Guarulhos University, Guarulhos, São Paulo, Brazil
| |
Collapse
|
30
|
Hanna JC, Corpas-Lopez V, Seizova S, Colon BL, Bacchetti R, Hall GMJ, Sands EM, Robinson L, Baragaña B, Wyllie S, Pawlowic MC. Mode of action studies confirm on-target engagement of lysyl-tRNA synthetase inhibitor and lead to new selection marker for Cryptosporidium. Front Cell Infect Microbiol 2023; 13:1236814. [PMID: 37600947 PMCID: PMC10436570 DOI: 10.3389/fcimb.2023.1236814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Cryptosporidiosis is a leading cause of diarrheal-associated morbidity and mortality, predominantly affecting children under 5 years old in low-and-middle-income countries. There is no effective treatment and no vaccine. New therapeutics are emerging from drug discovery efforts. It is critical that mode of action studies are performed alongside drug discovery to ensure the best clinical outcomes. Unfortunately, technology to identify and validate drug targets for Cryptosporidium is severely lacking. Methods We used C. parvum lysyl-tRNA synthetase (CpKRS) and DDD01510706 as a target-compound pair to develop both chemical and genetic tools for mode of action studies for Cryptosporidium. We adapted thermal proteome profiling (TPP) for Cryptosporidium, an unbiased approach for target identification. Results Using TPP we identified the molecular target of DDD01510706 and confirm that it is CpKRS. Genetic tools confirm that CpKRS is expressed throughout the life cycle and that this target is essential for parasite survival. Parasites genetically modified to over-express CpKRS or parasites with a mutation at the compound-binding site are resistant to treatment with DDD01510706. We leveraged these mutations to generate a second drug selection marker for genetic modification of Cryptosporidium, KRSR. This second selection marker is interchangeable with the original selection marker, NeoR, and expands the range of reverse genetic approaches available to study parasite biology. Due to the sexual nature of the Cryptosporidium life cycle, parental strains containing different drug selection markers can be crossed in vivo. Discussion Selection with both drug markers produces highly efficient genetic crosses (>99% hybrid progeny), paving the way for forward genetics approaches in Cryptosporidium.
Collapse
Affiliation(s)
- Jack C. Hanna
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Victor Corpas-Lopez
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Simona Seizova
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Beatrice L. Colon
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ross Bacchetti
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Grant M. J. Hall
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Emma M. Sands
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Lee Robinson
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Beatriz Baragaña
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Mattie C. Pawlowic
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
31
|
Lee S, Love MS, Modukuri R, Chatterjee AK, Huerta L, Lawson AP, McNamara CW, Mead JR, Hedstrom L, Cuny GD. Structure-activity relationship of BMS906024 derivatives for Cryptosporidium parvum growth inhibition. Bioorg Med Chem Lett 2023; 90:129328. [PMID: 37196868 PMCID: PMC10290938 DOI: 10.1016/j.bmcl.2023.129328] [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: 12/16/2022] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
BMS906024, a γ-secretase inhibitor that blocks Notch signaling, was previously shown to inhibit Cryptosporidium parvum growth in vitro. A structure-activity relationship (SAR) analysis of BMS906024 reported herein demonstrates the importance of the stereochemistry of the C-3 benzodiazepine and the succinyl β-substituent. However, concomitant removal of the succinyl α-substituent and switching the primary amide with secondary amides was tolerated. For example, 32 (SH287) inhibited C. parvum growth in HCT-8 host cells with an EC50 = 6.4 nM and an EC90 = 16 nM; however, blocking C. parvum growth with BMS906024 derivatives was correlative with inhibition of Notch signaling, highlighting that additional SAR analysis will be needed to separate these two activities.
Collapse
Affiliation(s)
- Seungheon Lee
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Health Building 2, Houston, TX 77204, USA
| | - Melissa S Love
- Calibr, a Division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ramkumar Modukuri
- Calibr, a Division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Arnab K Chatterjee
- Calibr, a Division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Lauren Huerta
- Calibr, a Division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ann P Lawson
- Department of Biology, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Case W McNamara
- Calibr, a Division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jan R Mead
- Atlanta VA Medical Center and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lizbeth Hedstrom
- Department of Biology, Brandeis University, 415 South St., Waltham, MA 02454, USA; Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Gregory D Cuny
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Health Building 2, Houston, TX 77204, USA.
| |
Collapse
|
32
|
Stanton C, Sun J, Nutsch K, Rosarda JD, Nguyen T, Li-Ma C, Kutseikin S, Saez E, Teijaro JR, Wiseman RL, Bollong MJ. Covalent targeting as a common mechanism for inhibiting NLRP3 inflammasome assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543248. [PMID: 37398499 PMCID: PMC10312593 DOI: 10.1101/2023.06.01.543248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The NLRP3 inflammasome is a cytosolic protein complex important for the regulation and secretion of inflammatory cytokines including IL-1β and IL-18. Aberrant overactivation of NLRP3 is implicated in numerous inflammatory disorders. However, the activation and regulation of NLRP3 inflammasome signaling remains poorly understood, limiting our ability to develop pharmacologic approaches to target this important inflammatory complex. Here, we developed and implemented a high-throughput screen to identify compounds that inhibit inflammasome assembly and activity. From this screen we identify and profile inflammasome inhibition of 20 new covalent compounds across 9 different chemical scaffolds, as well as many known inflammasome covalent inhibitors. Intriguingly, our results indicate that NLRP3 possesses numerous reactive cysteines on multiple domains whose covalent targeting blocks activation of this inflammatory complex. Specifically, focusing on compound VLX1570, which possesses multiple electrophilic moieties, we demonstrate that this compound allows covalent, intermolecular crosslinking of NLRP3 cysteines to inhibit inflammasome assembly. Our results, along with the recent identification of numerous covalent molecules that inhibit NLRP3 inflammasome activation, suggests that NLRP3 serves as a cellular electrophile sensor important for coordinating inflammatory signaling in response to redox stress. Further, our results support the potential for covalent cysteine modification of NLRP3 for regulating inflammasome activation and activity.
Collapse
Affiliation(s)
- Caroline Stanton
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jie Sun
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jessica D. Rosarda
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Thu Nguyen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Chloris Li-Ma
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Sergei Kutseikin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - John R. Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
33
|
Wang RS, Loscalzo J. Repurposing Drugs for the Treatment of COVID-19 and Its Cardiovascular Manifestations. Circ Res 2023; 132:1374-1386. [PMID: 37167362 PMCID: PMC10171294 DOI: 10.1161/circresaha.122.321879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
COVID-19 is an infectious disease caused by SARS-CoV-2 leading to the ongoing global pandemic. Infected patients developed a range of respiratory symptoms, including respiratory failure, as well as other extrapulmonary complications. Multiple comorbidities, including hypertension, diabetes, cardiovascular diseases, and chronic kidney diseases, are associated with the severity and increased mortality of COVID-19. SARS-CoV-2 infection also causes a range of cardiovascular complications, including myocarditis, myocardial injury, heart failure, arrhythmias, acute coronary syndrome, and venous thromboembolism. Although a variety of methods have been developed and many clinical trials have been launched for drug repositioning for COVID-19, treatments that consider cardiovascular manifestations and cardiovascular disease comorbidities specifically are limited. In this review, we summarize recent advances in drug repositioning for COVID-19, including experimental drug repositioning, high-throughput drug screening, omics data-based, and network medicine-based computational drug repositioning, with particular attention on those drug treatments that consider cardiovascular manifestations of COVID-19. We discuss prospective opportunities and potential methods for repurposing drugs to treat cardiovascular complications of COVID-19.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Channing Division of Network Medicine (R.-S.W., J.L.), Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School Boston, MA
| | - Joseph Loscalzo
- Channing Division of Network Medicine (R.-S.W., J.L.), Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School Boston, MA
- Division of Cardiovascular Medicine (J.L.), Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School Boston, MA
| |
Collapse
|
34
|
Caldwell N, Afshar R, Baragaña B, Bustinduy AL, Caffrey CR, Collins JJ, Fusco D, Garba A, Gardner M, Gomes M, Hoffmann KF, Hsieh M, Lo NC, McNamara CW, Nono JK, Padalino G, Read KD, Roestenberg M, Spangenberg T, Specht S, Gilbert IH. Perspective on Schistosomiasis Drug Discovery: Highlights from a Schistosomiasis Drug Discovery Workshop at Wellcome Collection, London, September 2022. ACS Infect Dis 2023; 9:1046-1055. [PMID: 37083395 PMCID: PMC10186373 DOI: 10.1021/acsinfecdis.3c00081] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Indexed: 04/22/2023]
Abstract
In September 2022, the Drug Discovery Unit at the University of Dundee, UK, organised an international meeting at the Wellcome Collection in London to explore the current clinical situation and challenges associated with treating schistosomiasis. The aim of this meeting was to discuss the need for new treatments in view of the clinical situation and to ascertain what the key requirements would be for any potential new anti-schistosomals. This information will be essential to inform ongoing drug discovery efforts for schistosomiasis. We also discussed the potential drug discovery pathway and associated criteria for progressing compounds to the clinic. To date, praziquantel (PZQ) is the only drug available to treat all species causing schistosomiasis, but it is often unable to completely clear parasites from an infected patient, partially due to its inactivity against juvenile worms. PZQ-mediated mass drug administration campaigns conducted in endemic areas (e.g., sub-Saharan Africa, where schistosomiasis is primarily prevalent) have contributed to reducing the burden of disease but will not eliminate the disease as a public health problem. The potential for Schistosoma to develop resistance towards PZQ, as the sole treatment available, could become a concern. Consequently, new anthelmintic medications are urgently needed, and this Perspective aims to capture some of the learnings from our discussions on the key criteria for new treatments.
Collapse
Affiliation(s)
- Nicola Caldwell
- Wellcome
Centre for Anti-Infectives Research, Drug Discovery Unit, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Rana Afshar
- Global
Health Institute of Merck, a subsidiary of Merck KGaA, Darmstadt,
Germany, Ares Trading
S.A., Route de Crassier 1, 1262 Eysins, Switzerland
| | - Beatriz Baragaña
- Wellcome
Centre for Anti-Infectives Research, Drug Discovery Unit, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Amaya L. Bustinduy
- Department
of Clinical Research, London School of Hygiene
& Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Conor R. Caffrey
- Center
for Discovery and Innovation in Parasitic Diseases, Skaggs School
of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC0657, La Jolla, California 92093, United States
| | - James J. Collins
- Department
of Pharmacology, UT Southwestern Medical
Center, Forest Park Road, Dallas, Texas 75235, United States
| | - Daniela Fusco
- Department
of Infectious Disease Epidemiology, Bernhard
Nocht Institute of Tropical Medicine, 20359 Hamburg, Germany
- German
Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, 38124 Brunswick, Germany
| | - Amadou Garba
- Department
of Control of Neglected Tropical Diseases, World Health Organization, 1202 Geneva, Switzerland
| | - Mark Gardner
- Salvensis
Ltd., 27 New Dover Rd., Canterbury, Kent CT1 3DN, United Kingdom
| | - Mireille Gomes
- Global
Health Institute of Merck, a subsidiary of Merck KGaA, Darmstadt,
Germany, Ares Trading
S.A., Route de Crassier 1, 1262 Eysins, Switzerland
| | - Karl F. Hoffmann
- Department
of Life Sciences (DLS), Aberystwyth University, Edward Llwyd Building, Aberystwyth SY23 3DA, United Kingdom
| | - Michael Hsieh
- Division
of Urology, Children’s National Hospital, and Department of
Urology, George Washington University, Washington, D.C. 20010, United States
| | - Nathan C. Lo
- Division
of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco, San Francisco, California 94110, United States
| | - Case W. McNamara
- Calibr,
a division of Scripps Research, North Torrey Pines Road, La Jolla, California 92037, United States
| | - Justin Komguep Nono
- Unit
of Immunobiology and Helminth Infections, Institute of Medical Research
and Medicinal Plant Studies (IMPM), Ministry
of Scientific Research and Innovation, Yaoundé 13033, Cameroon
| | - Gilda Padalino
- School
of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, United
Kingdom
| | - Kevin D. Read
- Wellcome
Centre for Anti-Infectives Research, Drug Discovery Unit, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Meta Roestenberg
- Department
of Parasitology and Department of Infectious Diseases, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Thomas Spangenberg
- Global
Health Institute of Merck, a subsidiary of Merck KGaA, Darmstadt,
Germany, Ares Trading
S.A., Route de Crassier 1, 1262 Eysins, Switzerland
| | - Sabine Specht
- Drugs for Neglected Diseases Initiative, 1202 Geneva, Switzerland
| | - Ian H. Gilbert
- Wellcome
Centre for Anti-Infectives Research, Drug Discovery Unit, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, United Kingdom
| |
Collapse
|
35
|
Padalino G, Coghlan A, Pagliuca G, Forde-Thomas JE, Berriman M, Hoffmann KF. Using ChEMBL to Complement Schistosome Drug Discovery. Pharmaceutics 2023; 15:1359. [PMID: 37242601 PMCID: PMC10220823 DOI: 10.3390/pharmaceutics15051359] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Schistosomiasis is one of the most important neglected tropical diseases. Until an effective vaccine is registered for use, the cornerstone of schistosomiasis control remains chemotherapy with praziquantel. The sustainability of this strategy is at substantial risk due to the possibility of praziquantel insensitive/resistant schistosomes developing. Considerable time and effort could be saved in the schistosome drug discovery pipeline if available functional genomics, bioinformatics, cheminformatics and phenotypic resources are systematically leveraged. Our approach, described here, outlines how schistosome-specific resources/methodologies, coupled to the open-access drug discovery database ChEMBL, can be cooperatively used to accelerate early-stage, schistosome drug discovery efforts. Our process identified seven compounds (fimepinostat, trichostatin A, NVP-BEP800, luminespib, epoxomicin, CGP60474 and staurosporine) with ex vivo anti-schistosomula potencies in the sub-micromolar range. Three of those compounds (epoxomicin, CGP60474 and staurosporine) also demonstrated potent and fast-acting ex vivo effects on adult schistosomes and completely inhibited egg production. ChEMBL toxicity data were also leveraged to provide further support for progressing CGP60474 (as well as luminespib and TAE684) as a novel anti-schistosomal compound. As very few compounds are currently at the advanced stages of the anti-schistosomal pipeline, our approaches highlight a strategy by which new chemical matter can be identified and quickly progressed through preclinical development.
Collapse
Affiliation(s)
- Gilda Padalino
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Avril Coghlan
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK;
| | | | | | - Matthew Berriman
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK;
| | - Karl F. Hoffmann
- The Department of Life Sciences (DLS), Aberystwyth University, Aberystwyth SY23 3DA, UK;
| |
Collapse
|
36
|
Chiem K, Nogales A, Lorenzo M, Vasquez DM, Xiang Y, Gupta YK, Blasco R, de la Torre JC, Mart Nez-Sobrido L. Antivirals against monkeypox infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537483. [PMID: 37131608 PMCID: PMC10153157 DOI: 10.1101/2023.04.19.537483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Monkeypox virus (MPXV) infection in humans are historically restricted to endemic regions in Africa. However, in 2022, an alarming number of MPXV cases have been reported globally with evidence of person-to-person transmission. Because of this, the World Health Organization (WHO) declared the MPXV outbreak a public health emergency of international concern. MPXV vaccines are limited and only two antivirals, tecovirimat and brincidofovir, approved by the United States (US) Food and Drug Administration (FDA) for the treatment of smallpox, are currently available for the treatment of MPXV infection. Here, we evaluated 19 compounds previously shown to inhibit different RNA viruses for their ability to inhibit Orthopoxvirus infections. We first used recombinant vaccinia virus (rVACV) expressing fluorescence (Scarlet or GFP) and luciferase (Nluc) reporter genes to identify compounds with anti-Orthopoxvirus activity. Seven compounds from the ReFRAME library (antimycin A, mycophenolic acid, AVN- 944, pyrazofurin, mycophenolate mofetil, azaribine, and brequinar) and six compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) showed antiviral activity against rVACV. Notably, the anti-VACV activity of some of the compounds in the ReFRAME library (antimycin A, mycophenolic acid, AVN- 944, mycophenolate mofetil, and brequinar) and all the compounds from the NPC library (buparvaquone, valinomycin, narasin, monensin, rotenone, and mubritinib) were confirmed with MPXV, demonstrating the broad-spectrum antiviral activity against Orthopoxviruses and their potential to be used for the antiviral treatment of MPXV, or other Orthopoxvirus, infections. IMPORTANCE Despite the eradication of smallpox, some Orthopoxviruses remain important human pathogens, as exemplified by the recent 2022 monkeypox virus (MPXV) outbreak. Although smallpox vaccines are effective against MPXV, there is presently limited access to those vaccines. In addition, current antiviral treatment against MPXV infections is limited to the use of the FDA-approved drugs tecovirimat and brincidofovir. Thus, there is an urgent need to identify novel antivirals for the treatment of MPXV, and other potentially zoonotic Orthopoxvirus infections. Here, we show that thirteen compounds, derived from two different libraries, previously found to inhibit several RNA viruses, exhibit also antiviral activity against VACV. Notably, eleven compounds also displayed antiviral activity against MPXV, demonstrating their potential to be incorporated into the therapeutic armamentarium to combat Orthopoxvirus infections.
Collapse
|
37
|
Carucci M, Duez J, Tarning J, García-Barbazán I, Fricot-Monsinjon A, Sissoko A, Dumas L, Gamallo P, Beher B, Amireault P, Dussiot M, Dao M, Hull MV, McNamara CW, Roussel C, Ndour PA, Sanz LM, Gamo FJ, Buffet P. Safe drugs with high potential to block malaria transmission revealed by a spleen-mimetic screening. Nat Commun 2023; 14:1951. [PMID: 37029122 PMCID: PMC10082216 DOI: 10.1038/s41467-023-37359-2] [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: 07/20/2022] [Accepted: 03/15/2023] [Indexed: 04/09/2023] Open
Abstract
Malaria parasites like Plasmodium falciparum multiply in red blood cells (RBC), which are cleared from the bloodstream by the spleen when their deformability is altered. Drug-induced stiffening of Plasmodium falciparum-infected RBC should therefore induce their elimination from the bloodstream. Here, based on this original mechanical approach, we identify safe drugs with strong potential to block the malaria transmission. By screening 13 555 compounds with spleen-mimetic microfilters, we identified 82 that target circulating transmissible form of P. falciparum. NITD609, an orally administered PfATPase inhibitor with known effects on P. falciparum, killed and stiffened transmission stages in vitro at nanomolar concentrations. Short exposures to TD-6450, an orally-administered NS5A hepatitis C virus inhibitor, stiffened transmission parasite stages and killed asexual stages in vitro at high nanomolar concentrations. A Phase 1 study in humans with a primary safety outcome and a secondary pharmacokinetics outcome ( https://clinicaltrials.gov , ID: NCT02022306) showed no severe adverse events either with single or multiple doses. Pharmacokinetic modelling showed that these concentrations can be reached in the plasma of subjects receiving short courses of TD-6450. This physiologically relevant screen identified multiple mechanisms of action, and safe drugs with strong potential as malaria transmission-blocking agents which could be rapidly tested in clinical trials.
Collapse
Affiliation(s)
- Mario Carucci
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | | | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 10400, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Irene García-Barbazán
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28222, Madrid, Spain
| | - Aurélie Fricot-Monsinjon
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Abdoulaye Sissoko
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Lucie Dumas
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Pablo Gamallo
- Global Health Medicines R&D, GlaxoSmith Kline (GSK), 28760, Tres Cantos, Spain
| | - Babette Beher
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Pascal Amireault
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
- Laboratory of cellular and molecular mechanisms of hematological disorders and therapeutic implications, INSERM, 75014, Paris, France
| | - Michael Dussiot
- Laboratory of cellular and molecular mechanisms of hematological disorders and therapeutic implications, INSERM, 75014, Paris, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, MA, 02139, Cambridge, USA
| | - Mitchell V Hull
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Camille Roussel
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Laboratoire d'Hématologie générale, Hôpital Universitaire Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015, Paris, France
| | - Papa Alioune Ndour
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Laura Maria Sanz
- Global Health Medicines R&D, GlaxoSmith Kline (GSK), 28760, Tres Cantos, Spain
| | | | - Pierre Buffet
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France.
- Department of Infectious & Tropical Disease, AP-HP, Necker Hospital, 75015, Paris, France.
- Centre Médical de l'Institut Pasteur (CMIP), Institut Pasteur, 75015, Paris, France.
| |
Collapse
|
38
|
Smith E, Davis-Gardner ME, Garcia-Ordonez RD, Nguyen TT, Hull M, Chen E, Yu X, Bannister TD, Baillargeon P, Scampavia L, Griffin P, Farzan M, Spicer TP. High throughput screening for drugs that inhibit 3C-like protease in SARS-CoV-2. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:95-101. [PMID: 36646172 PMCID: PMC9839384 DOI: 10.1016/j.slasd.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/14/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
The SARS coronavirus 2 (SARS-CoV-2) pandemic remains a major problem in many parts of the world and infection rates remain at extremely high levels. This high prevalence drives the continued emergence of new variants, and possibly ones that are more vaccine-resistant and that can drive infections even in highly vaccinated populations. The high rate of variant evolution makes clear the need for new therapeutics that can be clinically applied to minimize or eliminate the effects of COVID-19. With a hurdle of 10 years, on average, for first in class small molecule therapeutics to achieve FDA approval, the fastest way to identify therapeutics is by drug repurposing. To this end, we developed a high throughput cell-based screen that incorporates the essential viral 3C-like protease and its peptide cleavage site into a luciferase complementation assay to evaluate the efficacy of known drugs encompassing approximately 15,000 clinical-stage or FDA-approved small molecules. Confirmed inhibitors were also tested to determine their cytotoxic properties. Medicinal chemistry efforts to optimize the hits identified Tranilast as a potential lead. Here, we report the rapid screening and identification of potentially relevant drugs that exhibit selective inhibition of the SARS-CoV-2 viral 3C-like protease.
Collapse
Affiliation(s)
- Emery Smith
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | | | - Ruben D Garcia-Ordonez
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Tu-Trinh Nguyen
- Calibr at Scripps Research, La Jolla, CA 92037, United States
| | - Mitchell Hull
- Calibr at Scripps Research, La Jolla, CA 92037, United States
| | - Emily Chen
- Calibr at Scripps Research, La Jolla, CA 92037, United States
| | - Xuerong Yu
- Department of Chemistry, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Thomas D Bannister
- Department of Chemistry, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Pierre Baillargeon
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Louis Scampavia
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Patrick Griffin
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Michael Farzan
- Immunology and Microbiology, UF Scripps Biomedical Research, Jupiter, FL 33458, United States
| | - Timothy P Spicer
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, United States.
| |
Collapse
|
39
|
Gao H, Dai R, Su R. Computer-aided drug design for the pain-like protease (PL pro) inhibitors against SARS-CoV-2. Biomed Pharmacother 2023; 159:114247. [PMID: 36689835 PMCID: PMC9841087 DOI: 10.1016/j.biopha.2023.114247] [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: 11/25/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
A new coronavirus, known as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is a highly contagious virus and has caused a massive worldwide health crisis. While large-scale vaccination efforts are underway, the management of population health, economic impact and asof-yet unknown long-term effects on physical and mental health will be a key challenge for the next decade. The papain-like protease (PLpro) of SARS-CoV-2 is a promising target for antiviral drugs. This report used pharmacophore-based drug design technology to identify potential compounds as PLpro inhibitors against SARS-CoV-2. The optimal pharmacophore model was fully validated using different strategies and then was employed to virtually screen out 10 compounds with inhibitory. Molecular docking and non-bonding interactions between the targeted protein PLpro and compounds showed that UKR1129266 was the best compound. These results provided a theoretical foundation for future studies of PLpro inhibitors against SARS-CoV-2.
Collapse
Affiliation(s)
- Hongwei Gao
- School of Life Science, Ludong University, Yantai, Shandong 264025, China.
| | | | | |
Collapse
|
40
|
Miglianico M, Bolscher JM, Vos MW, Koolen KJM, de Bruijni M, Rajagopal DS, Chen E, Kiczun M, Gray D, Campo B, Sauerwein RW, Dechering KJ. Assessment of the drugability of initial malaria infection through miniaturized sporozoite assays and high-throughput screening. Commun Biol 2023; 6:216. [PMID: 36823266 PMCID: PMC9950425 DOI: 10.1038/s42003-023-04599-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
The sporozoite stages of malaria parasites are the primary cause of infection of the vertebrate host and are targeted by (experimental) vaccines. Yet, little is known about their susceptibility to chemical intervention. Phenotypic high-throughput screens have not been feasible due to a lack of in vitro systems. Here we tested 78 marketed and experimental antimalarial compounds in miniaturized assays addressing sporozoite viability, gliding motility, hepatocyte traversal, and intrahepatocytic schizogony. None potently interfered with sporozoite viability or motility but ten compounds acted at the level of schizogony with IC50s < 100 nM. To identify compounds directly targeting sporozoites, we screened 81,000 compounds from the Global Health Diversity and reFRAME libraries in a sporozoite viability assay using a parasite expressing a luciferase reporter driven by the circumsporozoite promoter. The ionophore gramicidin emerged as the single hit from this screening campaign. Its effect on sporozoite viability translated into reduced gliding motility and an inability of sporozoites to invade human primary hepatocytes and develop into hepatic schizonts. While providing proof of concept for a small molecule sporontocidal mode of action, our combined data indicate that liver schizogony is more accessible to chemical intervention by (candidate) antimalarials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emily Chen
- Calibr, a division of The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael Kiczun
- Drug Discovery Unit, University of Dundee, Dundee, United Kingdom
| | - David Gray
- Drug Discovery Unit, University of Dundee, Dundee, United Kingdom
| | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | | | | |
Collapse
|
41
|
Tyrosine Kinase Inhibitors Display Potent Activity against Cryptosporidium parvum. Microbiol Spectr 2023; 11:e0387422. [PMID: 36533912 PMCID: PMC9927415 DOI: 10.1128/spectrum.03874-22] [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] [Indexed: 12/24/2022] Open
Abstract
The protozoan parasite Cryptosporidium is a leading cause of diarrheal disease (cryptosporidiosis) and death in young children. Cryptosporidiosis can be life-threatening in individuals with weak immunity such as HIV/AIDS patients and organ transplant recipients. There is currently no effective drug to treat cryptosporidiosis in the pediatric and immunocompromised population. Therefore, there is an urgent need to expedite the drug discovery process in order to develop new and effective therapies to reduce the global disease burden of cryptosporidiosis. In this study, we employed a drug repurposing strategy to screen a library of 473 human kinase inhibitors to determine their activity against Cryptosporidium parvum. We have identified 67 new anti-cryptosporidial compounds using phenotypic screening based on a transgenic C. parvum strain expressing a luciferase reporter. Further, dose-response assays led to the identification of 11 hit compounds that showed potent inhibition of C. parvum at nanomolar concentration. Kinome profiling of these 11 prioritized hits identified compounds that displayed selectivity in targeting specific families of kinases, particularly tyrosine kinases. Overall, this study identified tyrosine kinase inhibitors that hold potential for future development as new drug candidates against cryptosporidiosis. IMPORTANCE The intestinal parasite Cryptosporidium parvum is a major cause of diarrhea-associated morbidity and mortality in children, immunocompromised people, and young ruminant animals. With no effective drug available to treat cryptosporidiosis in humans and animals, there is an urgent need to identify anti-parasitic compounds and new targets for drug development. To address this unmet need, we screened a GSK library of kinase inhibitors and identified several potent compounds, including tyrosine kinase inhibitors, that were highly effective in killing C. parvum. Overall, our study revealed several novel compounds and a new family of kinases that can be targeted for anti-cryptosporidial drug development.
Collapse
|
42
|
Ohzono H, Hu Y, Nagira K, Kanaya H, Okubo N, Olmer M, Gotoh M, Kurakazu I, Akasaki Y, Kawata M, Chen E, Chu AC, Johnson KA, Lotz MK. Targeting FoxO transcription factors with HDAC inhibitors for the treatment of osteoarthritis. Ann Rheum Dis 2023; 82:262-271. [PMID: 36109140 PMCID: PMC11005918 DOI: 10.1136/ard-2021-221269] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/27/2022] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Osteoarthritis (OA) features ageing-related defects in cellular homeostasis mechanisms in articular cartilage. These defects are associated with suppression of forkhead box O (FoxO) transcription factors. FoxO1 or FoxO3 deficient mice show early onset OA while FoxO1 protects against oxidative stress in chondrocytes and promotes expression of autophagy genes and the essential joint lubricant proteoglycan 4 (PRG4). The objective of this study was to identify small molecules that can increase FoxO1 expression. METHODS We constructed a reporter cell line with FoxO1 promoter sequences and performed high-throughput screening (HTS) of the Repurposing, Focused Rescue and Accelerated Medchem (ReFRAME) library . Hits from the HTS were validated and function was assessed in human chondrocytes, meniscus cells and synoviocytes and following administration to mice. The most promising hit, the histone deacetylase inhibitor (HDACI) panobinostat was tested in a murine OA model. RESULTS Among the top hits were HDACI and testing in human chondrocytes, meniscus cells and synoviocytes showed that panobinostat was the most promising compound as it increased the expression of autophagy genes and PRG4 while suppressing the basal and IL-1β induced expression of inflammatory mediators and extracellular matrix degrading enzymes. Intraperitoneal administration of panobinostat also suppressed the expression of mediators of OA pathogenesis induced by intra-articular injection of IL-1β. In a murine OA model, panobinostat reduced the severity of histological changes in cartilage, synovium and subchondral bone and improved pain behaviours. CONCLUSION Panobinostat has a clinically relevant activity profile and is a candidate for OA symptom and structure modification.
Collapse
Affiliation(s)
- Hiroki Ohzono
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Orthopaedic Surgery, Kurume University Hospital, Kurume, Japan
| | - Yiwen Hu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Radiology, Fudan University, Shanghai, China
| | - Keita Nagira
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Orthopaedic Surgery, Tottori University, Tottori, Japan
| | - Haruhisa Kanaya
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Orthopaedic Surgery, Tottori University, Tottori, Japan
| | - Naoki Okubo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Orthopaedics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Merissa Olmer
- The Scripps Research Institute, La Jolla, California, USA
| | - Masafumi Gotoh
- Department of Orthopaedic Surgery, Kurume University Hospital, Kurume, Japan
| | - Ichiro Kurakazu
- The Scripps Research Institute, La Jolla, California, USA
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Kyushu, Japan
| | - Yukio Akasaki
- Department of Orthopaedics, Kyushu University, Kyushu, UK
| | - Manabu Kawata
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Emily Chen
- Calibr, a Division of Scripps Research Institute, La Jolla, California, USA
| | - Alan C Chu
- Calibr, a Division of Scripps Research Institute, La Jolla, California, USA
| | - Kristen A Johnson
- Calibr, a Division of Scripps Research Institute, La Jolla, California, USA
| | - Martin K Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
43
|
Caravedo MA, White AC. Treatment of cryptosporidiosis: nitazoxanide yes, but we can do better. Expert Rev Anti Infect Ther 2023; 21:167-173. [PMID: 36533398 DOI: 10.1080/14787210.2023.2160704] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Cryptosporidiosis was initially recognized as an important cause of diarrhea in AIDS patients. It has been underdiagnosed in other populations. Recent studies have highlighted the importance of Cryptosporidium as a cause of diarrhea and malnutrition in young children in resource-poor countries and an emerging pathogen in organ-transplant recipients. AREAS COVERED Nitazoxanide is FDA approved for treatment of cryptosporidiosis in immunocompetent people. However, it is less effective in HIV and transplant patients and malnourished children. In transplant recipients, there is emerging data on antiparasitic combinations for cryptosporidiosis, including combinations of nitazoxanide, azithromycin, and in one case rifaximin. High-throughput phenotypic screens have identified some potential treatments. Among them, clofazimine was no better than placebo in a trial in AIDS patients. There have also been efforts to develop drug versus specific parasite targets. However, in part due to safety issues, none of these compounds have advanced into clinical trials. EXPERT OPINION Development of new and more efficacious therapies for cryptosporidium is imperative. Current approve therapy is far from optimal and lacks efficacy in high-risk populations, such as, patients living with HIV. Additionally, there is limited data on patients with other types of immunosuppression (Transplanted, autoimmune conditions, etc).
Collapse
Affiliation(s)
- Maria A Caravedo
- Infectious Disease Division Department of Internal Medicine University of Texas Medical Branch, Galveston, Texas, USA
| | - A Clinton White
- Infectious Disease Division Department of Internal Medicine University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
44
|
Johanssen T, McVeigh L, Erridge S, Higgins G, Straehla J, Frame M, Aittokallio T, Carragher NO, Ebner D. Glioblastoma and the search for non-hypothesis driven combination therapeutics in academia. Front Oncol 2023; 12:1075559. [PMID: 36733367 PMCID: PMC9886867 DOI: 10.3389/fonc.2022.1075559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma (GBM) remains a cancer of high unmet clinical need. Current standard of care for GBM, consisting of maximal surgical resection, followed by ionisation radiation (IR) plus concomitant and adjuvant temozolomide (TMZ), provides less than 15-month survival benefit. Efforts by conventional drug discovery to improve overall survival have failed to overcome challenges presented by inherent tumor heterogeneity, therapeutic resistance attributed to GBM stem cells, and tumor niches supporting self-renewal. In this review we describe the steps academic researchers are taking to address these limitations in high throughput screening programs to identify novel GBM combinatorial targets. We detail how they are implementing more physiologically relevant phenotypic assays which better recapitulate key areas of disease biology coupled with more focussed libraries of small compounds, such as drug repurposing, target discovery, pharmacologically active and novel, more comprehensive anti-cancer target-annotated compound libraries. Herein, we discuss the rationale for current GBM combination trials and the need for more systematic and transparent strategies for identification, validation and prioritisation of combinations that lead to clinical trials. Finally, we make specific recommendations to the preclinical, small compound screening paradigm that could increase the likelihood of identifying tractable, combinatorial, small molecule inhibitors and better drug targets specific to GBM.
Collapse
Affiliation(s)
- Timothy Johanssen
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Laura McVeigh
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Sara Erridge
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
| | - Geoffrey Higgins
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Joelle Straehla
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, United States
| | - Margaret Frame
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Neil O. Carragher
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel Ebner
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Albuquerque PC, Zicker F, Fonseca BP. Advancing drug repurposing research: Trends, collaborative networks, innovation and knowledge leaders. Drug Discov Today 2022; 27:103396. [DOI: 10.1016/j.drudis.2022.103396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/08/2022] [Accepted: 10/06/2022] [Indexed: 11/03/2022]
|
46
|
Bush JA, Meyer SM, Fuerst R, Tong Y, Li Y, Benhamou RI, Aikawa H, Zanon PRA, Gibaut QMR, Angelbello AJ, Gendron TF, Zhang YJ, Petrucelli L, Heick Jensen T, Childs-Disney JL, Disney MD. A blood-brain penetrant RNA-targeted small molecule triggers elimination of r(G 4C 2) exp in c9ALS/FTD via the nuclear RNA exosome. Proc Natl Acad Sci U S A 2022; 119:e2210532119. [PMID: 36409902 PMCID: PMC9860304 DOI: 10.1073/pnas.2210532119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
A hexanucleotide repeat expansion in intron 1 of the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia, or c9ALS/FTD. The RNA transcribed from the expansion, r(G4C2)exp, causes various pathologies, including intron retention, aberrant translation that produces toxic dipeptide repeat proteins (DPRs), and sequestration of RNA-binding proteins (RBPs) in RNA foci. Here, we describe a small molecule that potently and selectively interacts with r(G4C2)exp and mitigates disease pathologies in spinal neurons differentiated from c9ALS patient-derived induced pluripotent stem cells (iPSCs) and in two c9ALS/FTD mouse models. These studies reveal a mode of action whereby a small molecule diminishes intron retention caused by the r(G4C2)exp and allows the liberated intron to be eliminated by the nuclear RNA exosome, a multi-subunit degradation complex. Our findings highlight the complexity of mechanisms available to RNA-binding small molecules to alleviate disease pathologies and establishes a pipeline for the design of brain penetrant small molecules targeting RNA with novel modes of action in vivo.
Collapse
Affiliation(s)
- Jessica A. Bush
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Samantha M. Meyer
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Rita Fuerst
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Yuquan Tong
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Yue Li
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Raphael I. Benhamou
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Haruo Aikawa
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Patrick R. A. Zanon
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Quentin M. R. Gibaut
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Alicia J. Angelbello
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | | | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL32224
| | | | - Torben Heick Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus CDK-8000, Denmark
| | - Jessica L. Childs-Disney
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| | - Matthew D. Disney
- Department of Chemistry, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
- Department of Neuroscience, The Scripps Research Institute and UF Scripps Biomedical Research, University of Florida, Jupiter, FL33458
| |
Collapse
|
47
|
Phenotypic screening of the ReFRAME drug repurposing library to discover new drugs for treating sickle cell disease. Proc Natl Acad Sci U S A 2022; 119:e2210779119. [PMID: 36161945 PMCID: PMC9546543 DOI: 10.1073/pnas.2210779119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Stem cell transplantation and genetic therapies offer potential cures for patients with sickle cell disease (SCD), but these options require advanced medical facilities and are expensive. Consequently, these treatments will not be available for many years to the majority of patients suffering from this disease. What is urgently needed now is an inexpensive oral drug in addition to hydroxyurea, the only drug approved by the FDA that inhibits sickle-hemoglobin polymerization. Here, we report the results of the first phase of our phenotypic screen of the 12,657 compounds of the Scripps ReFRAME drug repurposing library using a recently developed high-throughput assay to measure sickling times following deoxygenation to 0% oxygen of red cells from sickle trait individuals. The ReFRAME library is a very important collection because the compounds are either FDA-approved drugs or have been tested in clinical trials. From dose-response measurements, 106 of the 12,657 compounds exhibit statistically significant antisickling at concentrations ranging from 31 nM to 10 μM. Compounds that inhibit sickling of trait cells are also effective with SCD cells. As many as 21 of the 106 antisickling compounds emerge as potential drugs. This estimate is based on a comparison of inhibitory concentrations with free concentrations of oral drugs in human serum. Moreover, the expected therapeutic potential for each level of inhibition can be predicted from measurements of sickling times for cells from individuals with sickle syndromes of varying severity. Our results should motivate others to develop one or more of these 106 compounds into drugs for treating SCD.
Collapse
|
48
|
Childs-Disney JL, Yang X, Gibaut QMR, Tong Y, Batey RT, Disney MD. Targeting RNA structures with small molecules. Nat Rev Drug Discov 2022; 21:736-762. [PMID: 35941229 PMCID: PMC9360655 DOI: 10.1038/s41573-022-00521-4] [Citation(s) in RCA: 200] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 01/07/2023]
Abstract
RNA adopts 3D structures that confer varied functional roles in human biology and dysfunction in disease. Approaches to therapeutically target RNA structures with small molecules are being actively pursued, aided by key advances in the field including the development of computational tools that predict evolutionarily conserved RNA structures, as well as strategies that expand mode of action and facilitate interactions with cellular machinery. Existing RNA-targeted small molecules use a range of mechanisms including directing splicing - by acting as molecular glues with cellular proteins (such as branaplam and the FDA-approved risdiplam), inhibition of translation of undruggable proteins and deactivation of functional structures in noncoding RNAs. Here, we describe strategies to identify, validate and optimize small molecules that target the functional transcriptome, laying out a roadmap to advance these agents into the next decade.
Collapse
Affiliation(s)
| | - Xueyi Yang
- Department of Chemistry, Scripps Research, Jupiter, FL, USA
| | | | - Yuquan Tong
- Department of Chemistry, Scripps Research, Jupiter, FL, USA
| | - Robert T Batey
- Department of Biochemistry, University of Colorado, Boulder, CO, USA.
| | | |
Collapse
|
49
|
Guijas C, To A, Montenegro-Burke JR, Domingo-Almenara X, Alipio-Gloria Z, Kok BP, Saez E, Alvarez NH, Johnson KA, Siuzdak G. Drug-Initiated Activity Metabolomics Identifies Myristoylglycine as a Potent Endogenous Metabolite for Human Brown Fat Differentiation. Metabolites 2022; 12:749. [PMID: 36005620 PMCID: PMC9415469 DOI: 10.3390/metabo12080749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Worldwide, obesity rates have doubled since the 1980s and in the USA alone, almost 40% of adults are obese, which is closely associated with a myriad of metabolic diseases such as type 2 diabetes and arteriosclerosis. Obesity is derived from an imbalance between energy intake and consumption, therefore balancing energy homeostasis is an attractive target for metabolic diseases. One therapeutic approach consists of increasing the number of brown-like adipocytes in the white adipose tissue (WAT). Whereas WAT stores excess energy, brown adipose tissue (BAT) can dissipate this energy overload in the form of heat, increasing energy expenditure and thus inhibiting metabolic diseases. To facilitate BAT production a high-throughput screening approach was developed on previously known drugs using human Simpson-Golabi-Behmel Syndrome (SGBS) preadipocytes. The screening allowed us to discover that zafirlukast, an FDA-approved small molecule drug commonly used to treat asthma, was able to differentiate adipocyte precursors and white-biased adipocytes into functional brown adipocytes. However, zafirlukast is toxic to human cells at higher dosages. Drug-Initiated Activity Metabolomics (DIAM) was used to investigate zafirlukast as a BAT inducer, and the endogenous metabolite myristoylglycine was then discovered to mimic the browning properties of zafirlukast without impacting cell viability. Myristoylglycine was found to be bio-synthesized upon zafirlukast treatment and was unique in inducing brown adipocyte differentiation, raising the possibility of using endogenous metabolites and bypassing the exogenous drugs to potentially alleviate disease, in this case, obesity and other related metabolic diseases.
Collapse
Affiliation(s)
- Carlos Guijas
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA 92037, USA
| | - Andrew To
- California Institute for Biomedical Research (Calibr), Scripps Research, La Jolla, CA 92037, USA
| | - J. Rafael Montenegro-Burke
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA 92037, USA
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Xavier Domingo-Almenara
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA 92037, USA
- Computational Metabolomics for Systems Biology Lab, Omics Sciences Unit, Eurecat—Technology Centre of Catalonia, 08005 Barcelona, Spain
| | - Zaida Alipio-Gloria
- California Institute for Biomedical Research (Calibr), Scripps Research, La Jolla, CA 92037, USA
| | - Bernard P. Kok
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Nicole H. Alvarez
- California Institute for Biomedical Research (Calibr), Scripps Research, La Jolla, CA 92037, USA
| | - Kristen A. Johnson
- California Institute for Biomedical Research (Calibr), Scripps Research, La Jolla, CA 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA 92037, USA
- Departments of Chemistry, Molecular, and Computational Biology, Scripps Research, La Jolla, CA 92037, USA
| |
Collapse
|
50
|
Dhal AK, Panda C, Yun SIL, Mahapatra RK. An update on Cryptosporidium biology and therapeutic avenues. J Parasit Dis 2022; 46:923-939. [PMID: 35755159 PMCID: PMC9215156 DOI: 10.1007/s12639-022-01510-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Cryptosporidium species has been identified as an important pediatric diarrheal pathogen in resource-limited countries, particularly in very young children (0–24 months). However, the only available drug (nitazoxanide) has limited efficacy and can only be prescribed in a medical setting to children older than one year. Many drug development projects have started to investigate new therapeutic avenues. Cryptosporidium’s unique biology is challenging for the traditional drug discovery pipeline and requires novel drug screening approaches. Notably, in recent years, new methods of oocyst generation, in vitro processing, and continuous three-dimensional cultivation capacities have been developed. This has enabled more physiologically pertinent research assays for inhibitor discovery. In a short time, many great strides have been made in the development of anti-Cryptosporidium drugs. These are expected to eventually turn into clinical candidates for cryptosporidiosis treatment in the future. This review describes the latest development in Cryptosporidium biology, genomics, transcriptomics of the parasite, assay development, and new drug discovery.
Collapse
Affiliation(s)
- Ajit Kumar Dhal
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Chinmaya Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Soon-IL Yun
- Department of Food Science and Technology, Jeonbuk National University, Jeonju, 54896 Republic of Korea
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896 Republic of Korea
| | | |
Collapse
|