1
|
Cruz-Bustos T, Eder T, Ruttkowski B, Joachim A. Deciphering the code of resistance: a genomic and transcriptomic exploration of the Cystoisospora suis Holland-I strain. Sci Rep 2025; 15:5461. [PMID: 39953090 PMCID: PMC11828913 DOI: 10.1038/s41598-025-89372-8] [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: 05/31/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Cystoisospora suis, a member of the apicomplexan order Coccidia and causative agent of neonatal porcine coccidiosis, poses a challenge to pig production due to the emergence of reduced efficacy of toltrazuril, the only EU-approved treatment. To address the critical gaps in understanding toltrazuril resistance and possibilities of early diagnostics, our study investigated the genetic basis of resistance through whole-genome DNA sequencing and transcriptome analysis of two C. suis strains, the toltrazuril-susceptible Wien-I and the resistant Holland-I. Additionally, we studied the mitochondrial genome and analysed mitochondrial gene expression in both strains. Our results show that genes encoding proteins involved in host-cell invasion displayed variable expression patterns and genetic mutations, suggesting adaptive changes in invasion mechanisms. Moreover, substantial fluctuations in the expression of genes linked to retrotransposons, accompanied by genetic alterations, were observed, highlighting their potential involvement in genomic rearrangements. Finally, our mitochondrial genome analyses revealed important insights into its genetic organization and conservation. Notably, the marked downregulation of CoI, CoIII and Cytb mRNA levels in the resistant strain Holland-I upon toltrazuril exposure highlights the dynamic response of mitochondrial genes to toltrazuril. These mitochondrial adaptations appear to be closely linked to the parasite drug resistance mechanism, potentially facilitating its survival under pharmacological stress. These findings enhance our knowledge of drug resistance mechanisms in Coccidia and highlight the need for novel management strategies, leading to the development of targeted treatments and controls.
Collapse
Affiliation(s)
- Teresa Cruz-Bustos
- Institute of Parasitology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria.
| | - Thomas Eder
- Institute for Medical Biochemistry, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Baerbel Ruttkowski
- Institute of Parasitology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria
| | - Anja Joachim
- Institute of Parasitology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria
| |
Collapse
|
2
|
Uo T, Ojo KK, Sprenger CC, Soriano Epilepsia K, Perera BGK, Damodarasamy M, Sun S, Kim S, Hogan HH, Hulverson MA, Choi R, Whitman GR, Barrett LK, Michaels SA, Xu LH, Sun VL, Arnold SL, Pang HJ, Nguyen MM, Vigil ALB, Kamat V, Sullivan LB, Sweet IR, Vidadala R, Maly DJ, Van Voorhis WC, Plymate SR. A Compound That Inhibits Glycolysis in Prostate Cancer Controls Growth of Advanced Prostate Cancer. Mol Cancer Ther 2024; 23:973-994. [PMID: 38507737 PMCID: PMC11219269 DOI: 10.1158/1535-7163.mct-23-0540] [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: 08/18/2023] [Revised: 12/19/2023] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Nonspecific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small-molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell-cycle, metabolic, and enzymatic assays were used to demonstrate their mechanism of action. A human patient-derived xenograft model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. We demonstrate a new class of small-molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Takuma Uo
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Kayode K. Ojo
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Cynthia C.T. Sprenger
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Kathryn Soriano Epilepsia
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - B. Gayani K. Perera
- Department of Chemistry, University of Washington; Seattle, Washington 98195, USA
| | - Mamatha Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Shihua Sun
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Soojin Kim
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Hannah H. Hogan
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Matthew A. Hulverson
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Ryan Choi
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Grant R. Whitman
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Lynn K. Barrett
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Samantha A. Michaels
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Linda H. Xu
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Vicky L. Sun
- Department of Pharmaceutics, University of Washington; Seattle, Washington 98195, USA
| | - Samuel L.M. Arnold
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
- Department of Pharmaceutics, University of Washington; Seattle, Washington 98195, USA
| | - Haley J. Pang
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Matthew M. Nguyen
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Anna-Lena B.G. Vigil
- Human Biology Division, Fred Hutchinson Cancer Center; Seattle, Washington 98109, USA
| | - Varun Kamat
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Diabetes Center, University of Washington; Seattle, Washington 98109, USA
| | - Lucas B. Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center; Seattle, Washington 98109, USA
- Department of Biochemistry, University of Washington; Seattle, Washington 98195, USA
| | - Ian R. Sweet
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Diabetes Center, University of Washington; Seattle, Washington 98109, USA
| | - Ram Vidadala
- Department of Chemistry, University of Washington; Seattle, Washington 98195, USA
| | - Dustin J. Maly
- Department of Chemistry, University of Washington; Seattle, Washington 98195, USA
| | - Wesley C. Van Voorhis
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Stephen R. Plymate
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System; Seattle, Washington 98108, USA
| |
Collapse
|
3
|
Uo T, Ojo KK, Sprenger CC, Epilepsia KS, Perera BGK, Damodarasamy M, Sun S, Kim S, Hogan HH, Hulverson MA, Choi R, Whitman GR, Barrett LK, Michaels SA, Xu LH, Sun VL, Arnold SLM, Pang HJ, Nguyen MM, Vigil ALBG, Kamat V, Sullivan LB, Sweet IR, Vidadala R, Maly DJ, Van Voorhis WC, Plymate SR. A Compound that Inhibits Glycolysis in Prostate Cancer Controls Growth of Advanced Prostate Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.547355. [PMID: 37461469 PMCID: PMC10350011 DOI: 10.1101/2023.07.01.547355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Purpose Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Non-specific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. Experimental design We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell cycle, metabolic and enzymatic assays were used to demonstrate their mechanism of action. A human PDX model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. Results We demonstrate a new class of small molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. Conclusion This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.
Collapse
|
4
|
Müller J, Hemphill A. In vitro screening technologies for the discovery and development of novel drugs against Toxoplasma gondii. Expert Opin Drug Discov 2024; 19:97-109. [PMID: 37921660 DOI: 10.1080/17460441.2023.2276349] [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: 08/31/2023] [Accepted: 10/24/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Toxoplasmosis constitutes a challenge for public health, animal production and welfare. Since more than 60 years, only a limited panel of drugs has been in use for clinical applications. AREAS COVERED Herein, the authors describe the methodology and the results of library screening approaches to identify inhibitors of Toxoplasma gondii and related strains. The authors then provide the reader with their expert perspectives for the future. EXPERT OPINION Various library screening projects, in particular those using reporter strains, have led to the identification of numerous compounds with good efficacy and specificity in vitro. However, only few compounds are effective in suitable animal models such as rodents. Whereas no novel compound has cleared the hurdle to applications in humans, the few compounds with known indication and application profiles in human patients are of interest for further investigations. Taken together, drug repurposing as well as high-throughput screening of novel compound libraries may shorten the way to novel drugs against toxoplasmosis.
Collapse
Affiliation(s)
- Joachim Müller
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Feix AS, Cruz-Bustos T, Ruttkowski B, Joachim A. In vitro cultivation methods for coccidian parasite research. Int J Parasitol 2023; 53:477-489. [PMID: 36400306 DOI: 10.1016/j.ijpara.2022.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022]
Abstract
The subclass Coccidia comprises a large group of protozoan parasites, including important pathogens of humans and animals such as Toxoplasma gondii, Neospora caninum, Eimeria spp., and Cystoisospora spp. Their life cycle includes a switch from asexual to sexual stages and is often restricted to a single host species. Current research on coccidian parasites focuses on cell biology and the underlying mechanisms of protein expression and trafficking in different life stages, host cell invasion and host-parasite interactions. Furthermore, novel anticoccidial drug targets are evaluated. Given the variety of research questions and the requirement to reduce and replace animal experimentation, in vitro cultivation of Coccidia needs to be further developed and refined to meet these requirements. For these purposes, established culture systems are constantly improved. In addition, new in vitro culture systems lately gained considerable importance in research on Coccidia. Well established and optimized in vitro cultures of monolayer cells can support the viability and development of parasite stages and even allow completion of the life cycle in vitro, as shown for Cystoisospora suis and Eimeria tenella. Furthermore, new three-dimensional cell culture models are used for propagation of Cryptosporidium spp. (close relatives of the coccidians), and the infection of three-dimensional organoids with T. gondii also gained popularity as the interaction between the parasite and host tissue can be studied in more detail. The latest advances in three-dimensional culture systems are organ-on-a-chip models, that to date have only been tested for T. gondii but promise to accelerate research in other coccidians. Lastly, the completion of the life cycle of C. suis and Cryptosporidium parvum was reported to continue in a host cell-free environment following the first occurrence of asexual stages. Such axenic cultures are becoming increasingly available and open new avenues for research on parasite life cycle stages and novel intervention strategies.
Collapse
Affiliation(s)
- Anna Sophia Feix
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria.
| | - Teresa Cruz-Bustos
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Bärbel Ruttkowski
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Anja Joachim
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| |
Collapse
|
6
|
Anghel N, Imhof D, Winzer P, Balmer V, Ramseier J, Haenggeli K, Choi R, Hulverson MA, Whitman GR, Arnold SL, Ojo KK, Van Voorhis WC, Doggett JS, Ortega-Mora LM, Hemphill A. Endochin-like quinolones (ELQs) and bumped kinase inhibitors (BKIs): Synergistic and additive effects of combined treatments against Neospora caninum infection in vitro and in vivo. Int J Parasitol Drugs Drug Resist 2021; 17:92-106. [PMID: 34482255 PMCID: PMC8416643 DOI: 10.1016/j.ijpddr.2021.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 12/30/2022]
Abstract
The apicomplexan parasite Neospora caninum is an important causative agent of congenital neosporosis, resulting in abortion, birth of weak offspring and neuromuscular disorders in cattle, sheep, and many other species. Among several compound classes that are currently being developed, two have been reported to limit the effects of congenital neosporosis: (i) bumped kinase inhibitors (BKIs) target calcium dependent protein kinase 1 (CDPK1), an enzyme that is encoded by an apicoplast-derived gene and found only in apicomplexans and plants. CDPK1 is essential for host cell invasion and egress; (ii) endochin-like quinolones (ELQs) are inhibitors of the cytochrome bc1 complex of the mitochondrial electron transport chain and thus inhibit oxidative phosphorylation. We here report on the in vitro and in vivo activities of BKI-1748, and of ELQ-316 and its respective prodrugs ELQ-334 and ELQ-422, applied either as single-compounds or ELQ-BKI-combinations. In vitro, BKI-1748 and ELQ-316, as well as BKI-1748 and ELQ-334, acted synergistically, while this was not observed for the BKI-1748/ELQ-422 combination treatment. In a N. caninum-infected pregnant BALB/c mouse model, the synergistic effects observed in vitro were not entirely reproduced, but 100% postnatal survival and 100% inhibition of vertical transmission was noted in the group treated with the BKI-1748/ELQ-334 combination. In addition, the combined drug applications resulted in lower neonatal mortality compared to treatments with single drugs.
Collapse
Affiliation(s)
- Nicoleta Anghel
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland,Corresponding author. Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland.
| | - Dennis Imhof
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Pablo Winzer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Vreni Balmer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Jessica Ramseier
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Kai Haenggeli
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Ryan Choi
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew A. Hulverson
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Grant R. Whitman
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Samuel L.M. Arnold
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA,Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Kayode K. Ojo
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA,Departments of Global Health and Microbiology, University of Washington, Seattle, WA, USA
| | - J. Stone Doggett
- VA Portland Health Care System, Research and Development Service, Portland, OR, USA
| | - Luis M. Ortega-Mora
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Spain
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Corresponding author.
| |
Collapse
|
7
|
Molina DA, Ramos GA, Zamora-Vélez A, Gallego-López GM, Rocha-Roa C, Gómez-Marin JE, Cortes E. In vitro evaluation of new 4-thiazolidinones on invasion and growth of Toxoplasma gondii. Int J Parasitol Drugs Drug Resist 2021; 16:129-139. [PMID: 34102589 PMCID: PMC8187164 DOI: 10.1016/j.ijpddr.2021.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 11/18/2022]
Abstract
Treatments for toxoplasmosis such as pyrimethamine have shown numerous side effects. It has been reported that the likelihood of relapse associated with pyrimethamine-based therapy in patients with HIV and toxoplasmic encephalitis (TE) can have significant implications, even for patients who often develop new lesions in areas of the brain previously free of infection. This led us to research for new agents against Toxoplasma gondii. Recent findings have shown the potent biological activity of 4-thiazolidinones. We proposed to design and synthesize a new series of 2-hydrazono-4-thiazolidinones derivatives to evaluate the in vitro growth inhibition effect on T. gondii. The growth rates of T. gondii tachyzoites in Human Foreskin Fibroblast (HFF) cell culture were identified by two in vitro methodologies. The first one was by fluorescence in which green fluorescent RH parasites and cherry-red fluorescent ME49 parasites were used. The second one was a colorimetric methodology using β-Gal parasites of the RH strain constitutively expressing the enzyme beta-galactosidase. The 4-thiazolidinone derivatives 1B, 2B and 3B showed growth inhibition at the same level of Pyrimethamine. These compounds showed IC50 values of 1B (0.468-0.952 μM), 2B (0.204-0.349 μM) and 3B (0.661-1.015 μM) against T. gondii. As a measure of cytotoxicity the compounds showed a TD50 values of: 1B (60 μM), 2B (206 μM) and 3B (125 μM). The in vitro assays and molecular modeling results suggest that these compounds could act as possible inhibitors of the Calcium-Dependent Protein Kinase 1 of T. gondii. Further, our results support the fact that of combining appropriate detection technologies, combinatorial chemistry and computational biology is a good strategy for efficient drug discovery. These compounds merit in vivo analysis for anti-parasitic drug detection.
Collapse
Affiliation(s)
- Diego A Molina
- Grupo GEPAMOL, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, 630004, Colombia.
| | | | - Alejandro Zamora-Vélez
- Grupo GEPAMOL, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, 630004, Colombia
| | - Gina M Gallego-López
- Morgridge Institute for Research, Madison, WI, 53715, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cristian Rocha-Roa
- Biophysics of Tropical Diseases, Max Planck Tandem Group, University of Antioquia, Medellin, 050010, Colombia
| | - Jorge Enrique Gómez-Marin
- Grupo GEPAMOL, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, 630004, Colombia
| | | |
Collapse
|
8
|
ApiCOWplexa 2019 - 5th International Meeting on Apicomplexan Parasites in Farm Animals. Int J Parasitol 2021; 50:345-347. [PMID: 32503686 DOI: 10.1016/j.ijpara.2020.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
9
|
Van Voorhis WC, Hulverson MA, Choi R, Huang W, Arnold SLM, Schaefer DA, Betzer DP, Vidadala RSR, Lee S, Whitman GR, Barrett LK, Maly DJ, Riggs MW, Fan E, Kennedy TJ, Tzipori S, Doggett JS, Winzer P, Anghel N, Imhof D, Müller J, Hemphill A, Ferre I, Sanchez-Sanchez R, Ortega-Mora LM, Ojo KK. One health therapeutics: Target-Based drug development for cryptosporidiosis and other apicomplexa diseases. Vet Parasitol 2021; 289:109336. [PMID: 33418437 PMCID: PMC8582285 DOI: 10.1016/j.vetpar.2020.109336] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022]
Abstract
This is a review of the development of bumped-kinase inhibitors (BKIs) for the therapy of One Health parasitic apicomplexan diseases. Many apicomplexan infections are shared between humans and livestock, such as cryptosporidiosis and toxoplasmosis, as well as livestock only diseases such as neosporosis. We have demonstrated proof-of-concept for BKI therapy in livestock models of cryptosporidiosis (newborn calves infected with Cryptosporidium parvum), toxoplasmosis (pregnant sheep infected with Toxoplasma gondii), and neosporosis (pregnant sheep infected with Neospora caninum). We discuss the potential uses of BKIs for the treatment of diseases caused by apicomplexan parasites in animals and humans, and the improvements that need to be made to further develop BKIs.
Collapse
Affiliation(s)
- Wesley C Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA.
| | - Matthew A Hulverson
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Ryan Choi
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Wenlin Huang
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Samuel L M Arnold
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Deborah A Schaefer
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Dana P Betzer
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Rama S R Vidadala
- Department of Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Sangun Lee
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, 01536, USA
| | - Grant R Whitman
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Lynn K Barrett
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle, WA, 98195, USA
| | - Michael W Riggs
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | | | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, 01536, USA
| | - J Stone Doggett
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Pablo Winzer
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Nicoleta Anghel
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Dennis Imhof
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Joachim Müller
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Ignacio Ferre
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Roberto Sanchez-Sanchez
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Luis Miguel Ortega-Mora
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Kayode K Ojo
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
10
|
Cystoisospora suis merozoite development assay for screening of drug efficacy in vitro. Exp Parasitol 2020; 220:108035. [PMID: 33189737 DOI: 10.1016/j.exppara.2020.108035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/02/2023]
Abstract
Cystoisospora suis is a common diarrheal pathogen of piglets and typically controlled by metaphylactic toltrazuril application. Recently, toltrazuril resistance has been reported in the field; however, both evaluation of toltrazuril efficacy against field isolates and the anticoccidial drug development for pigs is hampered by costs and labor of animal experimentation. Therefore an in vitro merozoite development assay was developed to evaluate the efficacy of compounds against C. suis in vitro. Monolayers of IPEC-1 cells were infected with sporozoites derived from oocysts of defined C. suis laboratory strains and the optimal infection dose as well as concentration, time point and duration of treatment were evaluated by quantitative real-time PCR. Cell cultures were treated with bumped kinase inhibitor (BKI) 1369 at different time points to evaluate the possibility to delineate effects on different developmental stages in vitro during invasion and early infection, and to determine different inhibitory concentrations (IC50, IC95). BKI 1369 had an IC50 of 35 nM and an IC95 of 350 nM. Dose- and duration-dependent efficacy was seen when developing stages were treated with BKI 1369 after infection (days 0-1, 2-3 and 2-5) but not when sporozoites were pre-incubated with BKI 1369 before infection. Efficacies of further BKIs were also evaluated at 200 nM. BKI 1318, 1708, 1748 and 1862 had an efficacy comparable to that of BKI 1369 (which is also effective in vivo). BKI 1862 showed a more pronounced loss of efficacy in lower concentrations than BKI 1369, signifying pharmacokinetic differences of similar compounds detectable in vitro. In addition, the effects of toltrazuril and its metabolites, toltrazuril sulfoxide and toltrazuril sulfone, on a toltrazuril sensitive and a resistant strain of C. suis were evaluated. Inhibition of merozoite growth in vitro by toltrazuril and its metabolites was dose-dependent only for toltrazuril. Clear differences were noted for the effect on a toltrazuril-sensitive vs. a resistant strain, indicating that this in vitro assay has the capacity to delineate susceptible from resistant strains in vitro. It could also be used to evaluate and compare the efficacy of novel compounds against C. suis and support the determination of the optimal time point of treatment in vivo.
Collapse
|
11
|
Shrestha A, Ruttkowski B, Greber P, Whitman GR, Hulverson MA, Choi R, Michaels SA, Ojo KK, Van Voorhis WC, Joachim A. Reduced treatment frequencies with bumped kinase inhibitor 1369 are effective against porcine cystoisosporosis. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:37-45. [PMID: 32861205 PMCID: PMC7442133 DOI: 10.1016/j.ijpddr.2020.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 11/14/2022]
Abstract
Bumped kinase inhibitors (BKIs) are a new class of antiprotozoal drugs that target calcium-dependent protein kinase 1 (CDPK1) in various apicomplexan parasites. A multiple dose regimen of BKI 1369 has been shown to be highly effective against Cystoisospora suis (syn. Isospora suis), the causative agent of neonatal porcine coccidiosis. However, multiple dosing may not be widely applicable in the field. The present study aimed to determine the efficacy of reduced treatment frequencies with BKI 1369 against porcine cystoisosporosis in vitro and in vivo. Pre-incubation of sporozoites with BKI 1369 completely failed to inhibit the infection in vitro unless treatment was prolonged post-infection. Notably, a single treatment of infected cell cultures 2 days post-infection (dpi) resulted in a significant reduction of merozoite replication. In an experimental infection model, treatment of suckling piglets experimentally infected with C. suis 2 and 4 dpi with 20 mg BKI 1369/kg body weight completely suppressed oocyst excretion. A single treatment on the day of infection or 2 dpi suppressed oocyst excretion in 50% and 82% of the piglets and reduced the quantitative excretion in those that shed oocysts by 95.2% and 98.4%, respectively. Moreover, a significant increase in body weight gain and reduced number of diarrhea days were observed in BKI 1369 treated piglets compared to the control piglets, irrespective of time points and frequencies of treatment. Given that reduced treatment frequencies with BKI 1369 are comparable in efficacy to repeated applications without any adverse effects, this could be considered as a practical therapeutic alternative against porcine cystoisosporosis. BKI 1369 does not target host cell invasion by C. suis sporozoites but replication of merozoites. Single treatment with BKI 1369 in parallel with experimental infection is not effective for the control of cystoisosporosis. Two doses of BKI 1369 at 2 and 4 dpi completely suppressed oocyst excretion in piglets experimentally infected with C. suis.
Collapse
Affiliation(s)
- Aruna Shrestha
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine, Veterinärplatz 1, A-1210, Vienna, Austria.
| | - Bärbel Ruttkowski
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine, Veterinärplatz 1, A-1210, Vienna, Austria
| | - Patricia Greber
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine, Veterinärplatz 1, A-1210, Vienna, Austria
| | - Grant R Whitman
- Center for Emerging and Reemerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Matthew A Hulverson
- Center for Emerging and Reemerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Ryan Choi
- Center for Emerging and Reemerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Samantha A Michaels
- Center for Emerging and Reemerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Kayode K Ojo
- Center for Emerging and Reemerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Wesley C Van Voorhis
- Center for Emerging and Reemerging Infectious Diseases, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA; Departments of Microbiology and Global Health, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Anja Joachim
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine, Veterinärplatz 1, A-1210, Vienna, Austria
| |
Collapse
|
12
|
Anghel N, Winzer PA, Imhof D, Müller J, Langa X, Rieder J, Barrett LK, Vidadala RSR, Huang W, Choi R, Hulverson MA, Whitman GR, Arnold SL, Van Voorhis WC, Ojo KK, Maly DJ, Fan E, Hemphill A. Comparative assessment of the effects of bumped kinase inhibitors on early zebrafish embryo development and pregnancy in mice. Int J Antimicrob Agents 2020; 56:106099. [PMID: 32707170 DOI: 10.1016/j.ijantimicag.2020.106099] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 01/30/2023]
Abstract
Bumped kinase inhibitors (BKIs) are effective against a variety of apicomplexan parasites. Fifteen BKIs with promising in vitro efficacy against Neospora caninum tachyzoites, low cytotoxicity in mammalian cells, and no toxic effects in non-pregnant BALB/c mice were assessed in pregnant mice. Drugs were emulsified in corn oil and were applied by gavage for 5 days. Five BKIs did not affect pregnancy, five BKIs exhibited ~15-35% neonatal mortality and five compounds caused strong effects (infertility, abortion, stillbirth and pup mortality). Additionally, the impact of these compounds on zebrafish (Danio rerio) embryo development was assessed by exposing freshly fertilised eggs to 0.2-50 μM of BKIs and microscopic monitoring of embryo development in a blinded manner for 4 days. We propose an algorithm that includes quantification of malformations and embryo deaths, and established a scoring system that allows the calculation of an impact score (Si) indicating at which concentrations BKIs visibly affect zebrafish embryo development. Comparison of the two models showed that for nine compounds no clear correlation between Si and pregnancy outcome was observed. However, the three BKIs affecting zebrafish embryos only at high concentrations (≥40 μM) did not impair mouse pregnancy at all, and the three compounds that inhibited zebrafish embryo development already at 0.2 μM showed detrimental effects in the pregnancy model. Thus, the zebrafish embryo development test has limited predictive value to foresee pregnancy outcome in BKI-treated mice. We conclude that maternal health-related factors such as cardiovascular, pharmacokinetic and/or bioavailability properties also contribute to BKI-pregnancy effects.
Collapse
Affiliation(s)
- Nicoleta Anghel
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland
| | - Pablo A Winzer
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland
| | - Dennis Imhof
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland
| | - Joachim Müller
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland
| | - Xavier Langa
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Baltzerstrasse 2, CH-3000 Bern, Switzerland
| | - Jessica Rieder
- Centre for Fish and Wildlife Health (FIWI), Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Lynn K Barrett
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | | | - Wenlin Huang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Ryan Choi
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Mathew A Hulverson
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Grant R Whitman
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Samuel L Arnold
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Wesley C Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Kayode K Ojo
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland.
| |
Collapse
|
13
|
Bumped Kinase Inhibitors as therapy for apicomplexan parasitic diseases: lessons learned. Int J Parasitol 2020; 50:413-422. [PMID: 32224121 DOI: 10.1016/j.ijpara.2020.01.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 11/24/2022]
Abstract
Bumped Kinase Inhibitors, targeting Calcium-dependent Protein Kinase 1 in apicomplexan parasites with a glycine gatekeeper, are promising new therapeutics for apicomplexan diseases. Here we will review advances, as well as challenges and lessons learned regarding efficacy, safety, and pharmacology that have shaped our selection of pre-clinical candidates.
Collapse
|