1
|
Ali M, Xu C, Wang J, Kulyar MFEA, Li K. Emerging therapeutic avenues against Cryptosporidium: A comprehensive review. Vet Parasitol 2024; 331:110279. [PMID: 39116547 DOI: 10.1016/j.vetpar.2024.110279] [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: 04/19/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Cryptosporidium is among the top causes of life-threatening diarrheal infection in public health and livestock sectors. Despite its high prevalence and economic importance, currently, there is no vaccine. Control of this protozoan is difficult due to the excretion of many resistant oocysts in the feces of the infected host, which contaminate the environment. Paromomycin shows inconsistent results and isn't considered a reliable therapy for cryptosporidiosis. Nitazoxanide (NTZ), the only FDA-approved drug against this parasite, is less productive in impoverished children and PLWHA (people living with HIV/AIDS). The absence of mitochondria and apicoplast, its unique location inside enterocytes separated by parasitophorous vacuole, and, most importantly, challenges in its genetic manipulations are some hurdles to the drug-discovery process. A library of compounds has been tested against Cryptosporidium during in vitro and in vivo trials. However, there has still not been sufficient success in finding the drug of choice against this parasite. Recent genome editing technologies based on CRISPR/Cas-9 have explored the functions of the vital genes by producing transgenic parasites that help to screen a collection of compounds to find target-specific drugs, provided the sufficient availability of in vitro culturing platforms, efficient transfection methods, and analytic techniques. The use of herbal remedies against Cryptosporidium is also an emerging area of interest with sufficient clinical success due to enhanced concern regarding anthelmintic resistance. Here, we highlighted present treatment options with their associated limitations, the use of genetic tools and natural products against it to find safe, effective, and inexpensive drugs to control the ever-increasing global burden of this disease.
Collapse
Affiliation(s)
- Munwar Ali
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Chang Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jia Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | | | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
2
|
Ibnidris A, Liaskos N, Eldem E, Gunn A, Streffer J, Gold M, Rea M, Teipel S, Gardiol A, Boccardi M. Facilitating the use of the target product profile in academic research: a systematic review. J Transl Med 2024; 22:693. [PMID: 39075460 PMCID: PMC11288132 DOI: 10.1186/s12967-024-05476-1] [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: 05/13/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The Target Product Profile (TPP) is a tool used in industry to guide development strategies by addressing user needs and fostering effective communication among stakeholders. However, they are not frequently used in academic research, where they may be equally useful. This systematic review aims to extract the features of accessible TPPs, to identify commonalities and facilitate their integration in academic research methodology. METHODS We searched peer-reviewed papers published in English developing TPPs for different products and health conditions in four biomedical databases. Interrater agreement, computed on random abstract and paper sets (Cohen's Kappa; percentage agreement with zero tolerance) was > 0.91. We interviewed experts from industry contexts to gain insight on the process of TPP development, and extracted general and specific features on TPP use and structure. RESULTS 138 papers were eligible for data extraction. Of them, 92% (n = 128) developed a new TPP, with 41.3% (n = 57) focusing on therapeutics. The addressed disease categories were diverse; the largest (47.1%, n = 65) was infectious diseases. Only one TPP was identified for several fields, including global priorities like dementia. Our analyses found that 56.5% of papers (n = 78) was authored by academics, and 57.8% of TPPs (n = 80) featured one threshold level of product performance. The number of TPP features varied widely across and within product types (n = 3-44). Common features included purpose/context of use, shelf life for drug stability and validation aspects. Most papers did not describe the methods used to develop the TPP. We identified aspects to be taken into account to build and report TPPs, as a starting point for more focused initiatives guiding use by academics. DISCUSSION TPPs are used in academic research mostly for infectious diseases and have heterogeneous features. Our extraction of key features and common structures helps to understand the tool and widen its use in academia. This is of particular relevance for areas of notable unmet needs, like dementia. Collaboration between stakeholders is key for innovation. Tools to streamline communication such as TPPs would support the development of products and services in academia as well as industry.
Collapse
Affiliation(s)
- Aliaa Ibnidris
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
- Neuroscience Institute, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Nektarios Liaskos
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
- European Infrastructure for Translational Medicine (EATRIS), Amsterdam, The Netherlands
| | - Ece Eldem
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
| | | | - Johannes Streffer
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Michael Gold
- AriLex Life Sciences LLC, 780 Elysian Way, Deerfield, IL, 60015, USA
| | | | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University of Medicine Rostock, Rostock, Germany
| | - Alejandra Gardiol
- European Infrastructure for Translational Medicine (EATRIS), Amsterdam, The Netherlands
- Queen Mary University of London, London, UK
| | - Marina Boccardi
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Gehlsheimer Str. 20, 18147, Rostock, Germany.
- Department of Psychosomatic Medicine and Psychotherapy, University of Medicine Rostock, Rostock, Germany.
| |
Collapse
|
3
|
Ali M, Xu C, Nawaz S, Ahmed AE, Hina Q, Li K. Anti-Cryptosporidial Drug-Discovery Challenges and Existing Therapeutic Avenues: A "One-Health" Concern. Life (Basel) 2024; 14:80. [PMID: 38255695 PMCID: PMC10820218 DOI: 10.3390/life14010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Cryptosporidiosis is the leading cause of life-threatening diarrheal infection, especially in infants. Oocysts contaminate the environment, and also, being a zoonotic disease, cryptosporidiosis is a threat to One Health. Nitazoxanide is the only FDA-approved drug, effective only in immunocompetent adults, and is not safe for infants. The absence of mitochondria and apicoplast, the presence of an electron-dense band (ED band), hindrances in its genetic and phenotypic manipulations, and its unique position inside the host cell are some challenges to the anti-cryptosporidial drug-discovery process. However, many compounds, including herbal products, have shown efficacy against Cryptosporidium during in vitro and in vivo trials. Still, the "drug of choice" against this protozoan parasite, especially in immunocompromised individuals and infants, has not yet been explored. The One-Health approach addresses this issue, focusing on the intersection of animal, human, and environmental health. The objective of this review is to provide knowledge about novel anti-cryptosporidial drug targets, available treatment options with associated limitations, and possible future shifts toward natural products to treat cryptosporidiosis. The current review is organized to address the treatment and prevention of cryptosporidiosis. An anti-cryptosporidial drug that is effective in immunocompromised individuals and infants is a necessity of our time.
Collapse
Affiliation(s)
- Munwar Ali
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.); (C.X.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chang Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.); (C.X.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia;
| | - Qazal Hina
- Department of Animal Nutrition, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan;
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.); (C.X.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
4
|
Fenwick M, Reers AR, Liu Y, Zigweid R, Sankaran B, Shin J, Hulverson MA, Hammerson B, Fernández Álvaro E, Myler PJ, Kaushansky A, Van Voorhis WC, Fan E, Staker BL. Identification of and Structural Insights into Hit Compounds Targeting N-Myristoyltransferase for Cryptosporidium Drug Development. ACS Infect Dis 2023; 9:1821-1833. [PMID: 37722671 PMCID: PMC10580320 DOI: 10.1021/acsinfecdis.3c00151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Indexed: 09/20/2023]
Abstract
Each year, approximately 50,000 children under 5 die as a result of diarrhea caused by Cryptosporidium parvum, a protozoan parasite. There are currently no effective drugs or vaccines available to cure or prevent Cryptosporidium infection, and there are limited tools for identifying and validating targets for drug or vaccine development. We previously reported a high throughput screening (HTS) of a large compound library against Plasmodium N-myristoyltransferase (NMT), a validated drug target in multiple protozoan parasite species. To identify molecules that could be effective against Cryptosporidium, we counter-screened hits from the Plasmodium NMT HTS against Cryptosporidium NMT. We identified two potential hit compounds and validated them against CpNMT to determine if NMT might be an attractive drug target also for Cryptosporidium. We tested the compounds against Cryptosporidium using both cell-based and NMT enzymatic assays. We then determined the crystal structure of CpNMT bound to Myristoyl-Coenzyme A (MyrCoA) and structures of ternary complexes with MyrCoA and the hit compounds to identify the ligand binding modes. The binding site architectures display different conformational states in the presence of the two inhibitors and provide a basis for rational design of selective inhibitors.
Collapse
Affiliation(s)
- Michael
K. Fenwick
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Alexandra R. Reers
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Yi Liu
- Department
of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Rachael Zigweid
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Banumathi Sankaran
- Berkeley
Center for Structural Biology, Advanced Light Source, Berkeley National
Laboratory, Berkeley, California 94720, United States
| | - Janis Shin
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Matthew A. Hulverson
- Center
for Emerging and Re-emerging Infectious Diseases, Division of Allergy
and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Bradley Hammerson
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | | | - Peter J. Myler
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
- Department
of Global Health, University of Washington, Seattle, Washington 98195, United States
- Department
of Pediatrics, University of Washington, Seattle, Washington 98195, United States
| | - Alexis Kaushansky
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
- Center
for Emerging and Re-emerging Infectious Diseases, Division of Allergy
and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington 98109, United States
- Department
of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Wesley C. Van Voorhis
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Emerging and Re-emerging Infectious Diseases, Division of Allergy
and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Erkang Fan
- Department
of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Bart L. Staker
- Seattle
Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| |
Collapse
|
5
|
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: 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: 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
|
6
|
Ogbuigwe P, Roberts JM, Knox MA, Heiser A, Pita A, Haack NA, Garcia-Ramirez J, Velathanthiri N, Biggs P, French NP, Hayman DTS. A novel, stain-free, natural auto-fluorescent signal, Sig M, identified from cytometric and transcriptomic analysis of infectivity of Cryptosporidium hominis and Cryptosporidium parvum. Front Cell Infect Microbiol 2023; 13:1178576. [PMID: 37284498 PMCID: PMC10239843 DOI: 10.3389/fcimb.2023.1178576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/28/2023] [Indexed: 06/08/2023] Open
Abstract
Cryptosporidiosis is a worldwide diarrheal disease caused by the protozoan Cryptosporidium. The primary symptom is diarrhea, but patients may exhibit different symptoms based on the species of the Cryptosporidium parasite they are infected with. Furthermore, some genotypes within species are more transmissible and apparently virulent than others. The mechanisms underpinning these differences are not understood, and an effective in vitro system for Cryptosporidium culture would help advance our understanding of these differences. Using COLO-680N cells, we employed flow cytometry and microscopy along with the C. parvum-specific antibody Sporo-Glo™ to characterize infected cells 48 h following an infection with C. parvum or C. hominis. The Cryptosporidium parvum-infected cells showed higher levels of signal using Sporo-Glo™ than C. hominis-infected cells, which was likely because Sporo-Glo™ was generated against C. parvum. We found a subset of cells from infected cultures that expressed a novel, dose-dependent auto-fluorescent signal that was detectable across a range of wavelengths. The population of cells that expressed this signal increased proportionately to the multiplicity of infection. The spectral cytometry results confirmed that the signature of this subset of host cells closely matched that of oocysts present in the infectious ecosystem, pointing to a parasitic origin. Present in both C. parvum and C. hominis cultures, we named this Sig M, and due to its distinct profile in cells from both infections, it could be a better marker for assessing Cryptosporidium infection in COLO-680N cells than Sporo-Glo™. We also noted Sig M's impact on Sporo-Glo™ detection as Sporo-Glo™ uses fluoroscein-isothiocynate, which is detected where Sig M also fluoresces. Lastly, we used NanoString nCounter® analysis to investigate the transcriptomic landscape for the two Cryptosporidium species, assessing the gene expression of 144 host and parasite genes. Despite the host gene expression being at high levels, the levels of putative intracellular Cryptosporidium gene expression were low, with no significant difference from controls, which could be, in part, explained by the abundance of uninfected cells present as determined by both Sporo-Glo™ and Sig M analyses. This study shows for the first time that a natural auto-fluorescent signal, Sig M, linked to Cryptosporidium infection can be detected in infected host cells without any fluorescent labeling strategies and that the COLO-680N cell line and spectral cytometry could be useful tools to advance the understanding of Cryptosporidium infectivity.
Collapse
Affiliation(s)
- Paul Ogbuigwe
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | | | - Matthew A. Knox
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Axel Heiser
- Animal Health Solutions, Hopkirk Research Institute, AgResearch Ltd., Massey University, Palmerston North, New Zealand
| | - Anthony Pita
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Neville A. Haack
- Animal Health Solutions, Hopkirk Research Institute, AgResearch Ltd., Massey University, Palmerston North, New Zealand
| | - Juan Carlos Garcia-Ramirez
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Niluka Velathanthiri
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Patrick J. Biggs
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - Nigel P. French
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| | - David T. S. Hayman
- School of Veterinary Science, Hopkirk Research Institute, Massey University, Palmerston North, New Zealand
| |
Collapse
|
7
|
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: 10] [Impact Index Per Article: 3.3] [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
|
8
|
Pane S, Putignani L. Cryptosporidium: Still Open Scenarios. Pathogens 2022; 11:pathogens11050515. [PMID: 35631036 PMCID: PMC9143492 DOI: 10.3390/pathogens11050515] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 01/27/2023] Open
Abstract
Cryptosporidiosis is increasingly identified as a leading cause of childhood diarrhea and malnutrition in both low-income and high-income countries. The strong impact on public health in epidemic scenarios makes it increasingly essential to identify the sources of infection and understand the transmission routes in order to apply the right prevention or treatment protocols. The objective of this literature review was to present an overview of the current state of human cryptosporidiosis, reviewing risk factors, discussing advances in the drug treatment and epidemiology, and emphasizing the need to identify a government system for reporting diagnosed cases, hitherto undervalued.
Collapse
Affiliation(s)
- Stefania Pane
- Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics, 00146 Rome, Italy;
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, 00146 Rome, Italy
- Correspondence:
| |
Collapse
|
9
|
Gupta Y, Goicoechea S, Pearce CM, Mathur R, Romero JG, Kwofie SK, Weyenberg MC, Daravath B, Sharma N, Poonam, Akala HM, Kanzok SM, Durvasula R, Rathi B, Kempaiah P. The emerging paradigm of calcium homeostasis as a new therapeutic target for protozoan parasites. Med Res Rev 2022; 42:56-82. [PMID: 33851452 DOI: 10.1002/med.21804] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/10/2020] [Accepted: 03/31/2021] [Indexed: 12/13/2022]
Abstract
Calcium channels (CCs), a group of ubiquitously expressed membrane proteins, are involved in many pathophysiological processes of protozoan parasites. Our understanding of CCs in cell signaling, organelle function, cellular homeostasis, and cell cycle control has led to improved insights into their structure and functions. In this article, we discuss CCs characteristics of five major protozoan parasites Plasmodium, Leishmania, Toxoplasma, Trypanosoma, and Cryptosporidium. We provide a comprehensive review of current antiparasitic drugs and the potential of using CCs as new therapeutic targets. Interestingly, previous studies have demonstrated that human CC modulators can kill or sensitize parasites to antiparasitic drugs. Still, none of the parasite CCs, pumps, or transporters has been validated as drug targets. Information for this review draws from extensive data mining of genome sequences, chemical library screenings, and drug design studies. Parasitic resistance to currently approved therapeutics is a serious and emerging threat to both disease control and management efforts. In this article, we suggest that the disruption of calcium homeostasis may be an effective approach to develop new anti-parasite drug candidates and reduce parasite resistance.
Collapse
Affiliation(s)
- Yash Gupta
- Infectious Diseases, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Steven Goicoechea
- Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Catherine M Pearce
- Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Raman Mathur
- Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Jesus G Romero
- Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Samuel K Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, West African Center for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| | - Matthew C Weyenberg
- Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Bharathi Daravath
- Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Neha Sharma
- Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi, India
| | - Poonam
- Department of Chemistry, Miranda House University Enclave, University of Delhi, Delhi, India
| | | | - Stefan M Kanzok
- Department of Biology, Loyola University Chicago, Chicago, Illinois, USA
| | - Ravi Durvasula
- Infectious Diseases, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Brijesh Rathi
- Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi, India
| | | |
Collapse
|
10
|
Oboh E, Schubert TJ, Teixeira JE, Stebbins EE, Miller P, Philo E, Thakellapalli H, Campbell SD, Griggs DW, Huston CD, Meyers MJ. Optimization of the Urea Linker of Triazolopyridazine MMV665917 Results in a New Anticryptosporidial Lead with Improved Potency and Predicted hERG Safety Margin. J Med Chem 2021; 64:11729-11745. [PMID: 34342443 DOI: 10.1021/acs.jmedchem.1c01136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cryptosporidiosis is caused by infection of the small intestine by Cryptosporidium parasites, resulting in severe diarrhea, dehydration, malabsorption, and potentially death. The only FDA-approved therapeutic is only partially effective in young children and ineffective for immunocompromised patients. Triazolopyridazine MMV665917 is a previously reported anti-Cryptosporidium screening hit with in vivo efficacy but suffers from modest inhibition of the hERG ion channel, which could portend cardiotoxicity. Herein, we describe our initial development of structure-activity relationships of this novel lead series with a particular focus on optimization of the piperazine-urea linker. We have discovered that piperazine-acetamide is a superior linker resulting in identification of SLU-2633, which has an EC50 of 0.17 μM, an improved projected margin versus hERG, prolonged pharmacokinetic exposure in small intestine, and oral efficacy in vivo with minimal systemic exposure. SLU-2633 represents a significant advancement toward the identification of a new effective and safe treatment for cryptosporidiosis.
Collapse
Affiliation(s)
- Edmund Oboh
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
| | - Tanner J Schubert
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
| | - Jose E Teixeira
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont 05401, United States
| | - Erin E Stebbins
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont 05401, United States
| | - Peter Miller
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont 05401, United States
| | - Emily Philo
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
| | - Haresh Thakellapalli
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
| | - Scott D Campbell
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104, United States
| | - David W Griggs
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104, United States.,Institute for Drug and Biotherapeutic Innovation, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Christopher D Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont 05401, United States
| | - Marvin J Meyers
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States.,Institute for Drug and Biotherapeutic Innovation, Saint Louis University, St. Louis, Missouri 63103, United States
| |
Collapse
|
11
|
Huston CD. The Clofazimine for Treatment of Cryptosporidiosis in HIV-Infected Adults (CRYPTOFAZ) and Lessons Learned for Anticryptosporidial Drug Development. Clin Infect Dis 2021; 73:192-194. [PMID: 32277815 PMCID: PMC8427724 DOI: 10.1093/cid/ciaa425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Indexed: 11/12/2022] Open
Affiliation(s)
- Christopher D Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
12
|
Opportunities and Challenges in Developing a Cryptosporidium Controlled Human Infection Model for Testing Antiparasitic Agents. ACS Infect Dis 2021; 7:959-968. [PMID: 33822577 PMCID: PMC8154424 DOI: 10.1021/acsinfecdis.1c00057] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cryptosporidiosis is a leading cause of moderate-to-severe diarrhea in low- and middle-income countries, responsible for high mortality in children younger than two years of age, and it is also strongly associated with childhood malnutrition and growth stunting. There is no vaccine for cryptosporidiosis and existing therapeutic options are suboptimal to prevent morbidity and mortality in young children. Recently, novel therapeutic agents have been discovered through high-throughput phenotypic and target-based screening strategies, repurposing malaria hits, etc., and these agents have a promising preclinical in vitro and in vivo anti-Cryptosporidium efficacy. One key step in bringing safe and effective new therapies to young vulnerable children is the establishment of some prospect of direct benefit before initiating pediatric clinical studies. A Cryptosporidium controlled human infection model (CHIM) in healthy adult volunteers can be a robust clinical proof of concept model for evaluating novel therapeutics. CHIM could potentially accelerate the development path to pediatric studies by establishing the safety of a proposed pediatric dosing regimen and documenting preliminary efficacy in adults. We present, here, perspectives regarding the opportunities and perceived challenges with the Cryptosporidium human challenge model.
Collapse
|
13
|
Abstract
It is estimated that more than 1 billion people across the world are affected by a neglected tropical disease (NTD) that requires medical intervention. These diseases tend to afflict people in areas with high rates of poverty and cost economies billions of dollars every year. Collaborative drug discovery efforts are required to reduce the burden of these diseases in endemic regions. The release of “Open Access Boxes” is an initiative launched by Medicines for Malaria Venture (MMV) in collaboration with its partners to catalyze new drug discovery in neglected diseases. These boxes are mainly requested by biology researchers across the globe who may not otherwise have access to compounds to screen nor knowledge of the workflow that needs to be followed after identification of actives from their screening campaigns. Here, we present guidelines on how to move such actives beyond the hit identification stage, to help in capacity strengthening and enable a greater impact of the initiative.
Collapse
|
14
|
Ashigbie PG, Shepherd S, Steiner KL, Amadi B, Aziz N, Manjunatha UH, Spector JM, Diagana TT, Kelly P. Use-case scenarios for an anti-Cryptosporidium therapeutic. PLoS Negl Trop Dis 2021; 15:e0009057. [PMID: 33705395 PMCID: PMC7951839 DOI: 10.1371/journal.pntd.0009057] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cryptosporidium is a widely distributed enteric parasite that has an increasingly appreciated pathogenic role, particularly in pediatric diarrhea. While cryptosporidiosis has likely affected humanity for millennia, its recent "emergence" is largely the result of discoveries made through major epidemiologic studies in the past decade. There is no vaccine, and the only approved medicine, nitazoxanide, has been shown to have efficacy limitations in several patient groups known to be at elevated risk of disease. In order to help frontline health workers, policymakers, and other stakeholders translate our current understanding of cryptosporidiosis into actionable guidance to address the disease, we sought to assess salient issues relating to clinical management of cryptosporidiosis drawing from a review of the literature and our own field-based practice. This exercise is meant to help inform health system strategies for improving access to current treatments, to highlight recent achievements and outstanding knowledge and clinical practice gaps, and to help guide research activities for new anti-Cryptosporidium therapies.
Collapse
Affiliation(s)
- Paul G. Ashigbie
- Novartis Institute for Tropical Diseases, Emeryville, California, United States of America
| | - Susan Shepherd
- Alliance for International Medical Action (ALIMA), Dakar, Senegal
| | - Kevin L. Steiner
- The Ohio State University, Columbus, Ohio, United States of America
| | - Beatrice Amadi
- Children’s Hospital, University Teaching Hospitals, Lusaka, Zambia
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Natasha Aziz
- Novartis Institute for Tropical Diseases, Emeryville, California, United States of America
| | - Ujjini H. Manjunatha
- Novartis Institute for Tropical Diseases, Emeryville, California, United States of America
| | - Jonathan M. Spector
- Novartis Institute for Tropical Diseases, Emeryville, California, United States of America
| | - Thierry T. Diagana
- Novartis Institute for Tropical Diseases, Emeryville, California, United States of America
| | - Paul Kelly
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
15
|
Bellini V, Swale C, Brenier-Pinchart MP, Pezier T, Georgeault S, Laurent F, Hakimi MA, Bougdour A. Target Identification of an Antimalarial Oxaborole Identifies AN13762 as an Alternative Chemotype for Targeting CPSF3 in Apicomplexan Parasites. iScience 2020; 23:101871. [PMID: 33336164 PMCID: PMC7733022 DOI: 10.1016/j.isci.2020.101871] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/27/2020] [Accepted: 11/22/2020] [Indexed: 12/22/2022] Open
Abstract
Boron-containing compounds represent a promising class of molecules with proven efficacy against a wide range of pathogens, including apicomplexan parasites. Following lead optimization, the benzoxaborole AN13762 was identified as a preclinical candidate against the human malaria parasite, yet the molecular target remained uncertain. Here, we uncovered the parasiticidal mechanisms of AN13762, by combining forward genetics with transcriptome sequencing and computational mutation discovery and using Toxoplasma gondii as a relevant model for Apicomplexa. AN13762 was shown to target TgCPSF3, the catalytic subunit of the pre-mRNA cleavage and polyadenylation complex, as the anti-pan-apicomplexan benzoxaborole compound, AN3661. However, unique mutations within the TgCPSF3 catalytic site conferring resistance to AN13762 do not confer cross-protection against AN3661, suggesting a divergent resistance mechanism. Finally, in agreement with the high sequence conservation of CPSF3 between Toxoplasma and Cryptosporidium, AN13762 shows oral efficacy in cryptosporidiosis mouse model, a disease for which new drug development is of high priority.
Collapse
Affiliation(s)
- Valeria Bellini
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Christopher Swale
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Marie-Pierre Brenier-Pinchart
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Tiffany Pezier
- INRAE, Université François Rabelais de Tours, Centre Val de Loire, UMR1282 ISP, Laboratoire Apicomplexes et Immunité Mucosale, 37380 Nouzilly, France
| | - Sonia Georgeault
- Plateforme des Microscopies, Université et CHRU de Tours, 37000 Tours, France
| | - Fabrice Laurent
- INRAE, Université François Rabelais de Tours, Centre Val de Loire, UMR1282 ISP, Laboratoire Apicomplexes et Immunité Mucosale, 37380 Nouzilly, France
| | - Mohamed-Ali Hakimi
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Alexandre Bougdour
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
16
|
Vinayak S. Recent advances in genetic manipulation of Cryptosporidium. Curr Opin Microbiol 2020; 58:146-152. [PMID: 33161368 DOI: 10.1016/j.mib.2020.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 01/06/2023]
Abstract
Cryptosporidium is a leading cause of diarrhea-associated morbidity and mortality in young children. Currently, there is no fully effective drug to treat cryptosporidiosis and a complete lack of vaccine to prevent disease. For a long time, progress in the field of Cryptosporidium research has been hindered due to unavailability of methods to propagate the parasite, lack of efficient animal infection models and most importantly, the absence of technology to genetically manipulate the parasite. The recent advent of molecular genetics has been transformative for Cryptosporidium research, and is facilitating our fundamental understanding of parasite biology, and accelerating the pace of drug discovery. This review summarizes recent advancements in genetic manipulation and its applications for studying parasite gene function, host-parasite interactions and discovery of anti-cryptosporidial drugs.
Collapse
Affiliation(s)
- Sumiti Vinayak
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States.
| |
Collapse
|
17
|
Elmahallawy EK, Elshopakey GE, Saleh AA, Agil A, El-Morsey A, EL-shewehy DMM, Sad AS, Yanai T, Abdo W. S-Methylcysteine (SMC) Ameliorates Intestinal, Hepatic, and Splenic Damage Induced by Cryptosporidium parvum Infection Via Targeting Inflammatory Modulators and Oxidative Stress in Swiss Albino Mice. Biomedicines 2020; 8:biomedicines8100423. [PMID: 33076496 PMCID: PMC7602666 DOI: 10.3390/biomedicines8100423] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/03/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023] Open
Abstract
Cryptosporidiosis has been proposed to be one of the major causes of diarrhoeal disease in humans worldwide that possesses zoonotic concern. Thereby, this study investigated the potential effects of s-Methylcysteine (SMC) on the parasite in vivo followed by the measurement of cytokines, oxidative stress parameters, and an investigation of the major histopathological changes. Sixty male Swiss albino mice weighing 20-25 g were allocated equally into five groups and orally administered saline only (control), SMC only (SMC50) (50 mg/kg b.w.), and 104Cryptosporidium parvum oocysts per mouse via an esophageal tube (C + ve untreated). The fourth and fifth groups (C + SMC25, C + SMC50) administrated 104C. parvum oocysts combined with SMC25 (low dose) and 50 (high dose) mg/kg b.w., respectively. At days 7 and 14 post-infection (PI), the feces was collected from each group in order to count C. parvum oocysts. After two weeks of treatment, the animals were euthanized and the serum was collected for biochemical analysis. Next, the intestinal, spleen, and liver sections were dissected for histopathological examination. The results revealed lower oocyst numbers in the C + SMC25 and C + SMC50 groups compared to the infected untreated group. Moreover, higher doses of SMC treatment significantly reduced the enteritis induced by C. parvum in a dose-dependent manner. The hepatic lesions were also mitigated as demonstrated in C + SMC25 and C + SMC50 groups unlike the infected group via lowering the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) enzymes and increasing albumin and globulin serum levels. SMC administration also reduced cytokines production (SAP, TNF-α, IL-6, and IFN-γ) mediated by Cryptosporidium infection in contrast to the infected untreated group. There were marked lymphoid depletion and amyloidosis observed in the infected untreated group, while the treated groups showed obvious increase in the lymphoid elements. Moreover, the scoring of intestinal parasites, hepatic, and splenic lesions in the SMC-treated groups exhibited significantly lower pathological lesions in different organs in a dose-dependent manner, compared to the infected untreated group. Our results also revealed a significant change in the malondialdehyde content with an elevation of glutathione and superoxide dismutase in the intestines collected from C + SMC25 and C + SMC50 mice relative to the untreated group. Taken together, our results indicated that SMC could be a promising effective compound for treating and declining C. parvum infestation via restoring structural alterations in different tissues, enhancing antioxidant enzymes, and suppressing the cytokines liberation.
Collapse
Affiliation(s)
- Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
- Department of Biomedical Sciences, University of León (ULE), 24071 León, Spain
| | - Gehad E. Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Amira A. Saleh
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Ahmad Agil
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Ahmed El-Morsey
- Parasitology and Animal Diseases Department, Veterinary Research Division, National Research Centre, 33 El Buhouth St. (former El-Tahrir St.), Dokki, P.O., Giza 18010, Egypt;
| | | | - Ahmed S. Sad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University, Port Fuad 42526, Egypt;
| | - Tokuma Yanai
- Laboratory of Wildlife and Forensic Pathology, Biomedical Science Examination and Research Center, Department of Veterinary Medicine, Faculty of Veterinary Medicine, Okayama University of Science, 1-3, Ikoinooka, Imabari 794-0085, Ehime, Japan
- Correspondence:
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr el-Sheikh 33516, Egypt;
| |
Collapse
|
18
|
Vinayak S, Jumani RS, Miller P, Hasan MM, McLeod BI, Tandel J, Stebbins EE, Teixeira JE, Borrel J, Gonse A, Zhang M, Yu X, Wernimont A, Walpole C, Eckley S, Love MS, McNamara CW, Sharma M, Sharma A, Scherer CA, Kato N, Schreiber SL, Melillo B, Striepen B, Huston CD, Comer E. Bicyclic azetidines kill the diarrheal pathogen Cryptosporidium in mice by inhibiting parasite phenylalanyl-tRNA synthetase. Sci Transl Med 2020; 12:eaba8412. [PMID: 32998973 PMCID: PMC8381743 DOI: 10.1126/scitranslmed.aba8412] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Cryptosporidium is a protozoan parasite and a leading cause of diarrheal disease and mortality in young children. Currently, there are no fully effective treatments available to cure infection with this diarrheal pathogen. In this study, we report a broad drug repositioning effort that led to the identification of bicyclic azetidines as a new anticryptosporidial series. Members of this series blocked growth in in vitro culture of three Cryptosporidium parvum isolates with EC50 's in 1% serum of <0.4 to 96 nM, had comparable potencies against Cryptosporidium hominis and C. parvum, and was effective in three of four highly susceptible immunosuppressed mice with once-daily dosing administered for 4 days beginning 2 weeks after infection. Comprehensive genetic, biochemical, and chemical studies demonstrated inhibition of C. parvum phenylalanyl-tRNA synthetase (CpPheRS) as the mode of action of this new lead series. Introduction of mutations directly into the C. parvum pheRS gene by CRISPR-Cas9 genome editing resulted in parasites showing high degrees of compound resistance. In vitro, bicyclic azetidines potently inhibited the aminoacylation activity of recombinant ChPheRS. Medicinal chemistry optimization led to the identification of an optimal pharmacokinetic/pharmacodynamic profile for this series. Collectively, these data demonstrate that bicyclic azetidines are a promising series for anticryptosporidial drug development and establish a broad framework to enable target-based drug discovery for this infectious disease.
Collapse
Affiliation(s)
- Sumiti Vinayak
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Rajiv S Jumani
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Peter Miller
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Muhammad M Hasan
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Briana I McLeod
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jayesh Tandel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erin E Stebbins
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jose E Teixeira
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Julien Borrel
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA
| | - Arthur Gonse
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA
| | - Mingliang Zhang
- International Discovery Service Unit, WuXi AppTec (Tianjin) Co. Ltd., Tianjin 300457, P.R. China
| | - Xianshui Yu
- International Discovery Service Unit, WuXi AppTec (Tianjin) Co. Ltd., Tianjin 300457, P.R. China
| | - Amy Wernimont
- Structural Genomics Consortium, MaRS Building, South Tower, 101 College Street, Suite 700, Toronto, Ontario M5G 1L7, Canada
| | - Chris Walpole
- Structural Genomics Consortium, MaRS Building, South Tower, 101 College Street, Suite 700, Toronto, Ontario M5G 1L7, Canada
| | | | - Melissa S Love
- 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
| | - Manmohan Sharma
- Structural Parasitology, Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Road, New Delhi 110070, India
| | - Amit Sharma
- Structural Parasitology, Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Road, New Delhi 110070, India
| | - Christina A Scherer
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA
| | - Nobutaka Kato
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Bruno Melillo
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Christopher D Huston
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA.
| | - Eamon Comer
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
19
|
Love MS, McNamara CW. Phenotypic screening techniques for Cryptosporidium drug discovery. Expert Opin Drug Discov 2020; 16:59-74. [PMID: 32892652 DOI: 10.1080/17460441.2020.1812577] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Two landmark epidemiological studies identified Cryptosporidium spp. as a significant cause of diarrheal disease in pediatric populations in resource-limited countries. Notably, nitazoxanide is the only approved drug for treatment of cryptosporidiosis but shows limited efficacy. As a result, many drug discovery efforts have commenced to find improved treatments. The unique biology of Cryptosporidium presents challenges for traditional drug discovery methods, which has inspired new assay platforms to study parasite biology and drug screening. Areas covered: The authors review historical advancements in phenotypic-based assays and techniques for Cryptosporidium drug discovery, as well as recent advances that will define future drug discovery. The reliance on phenotypic-based screens and repositioning of phenotypic hits from other pathogens has quickly created a robust pipeline of potential cryptosporidiosis therapeutics. The latest advances involve new in vitro culture methods for oocyst generation, continuous culturing capabilities, and more physiologically relevant assays for testing compounds. Expert opinion: Previous phenotypic screening techniques have laid the groundwork for recent cryptosporidiosis drug discovery efforts. The resulting improved methodologies characterize compound activity, identify, and validate drug targets, and prioritize new compounds for drug development. The most recent improvements in phenotypic assays are poised to help advance compounds into clinical development.
Collapse
Affiliation(s)
- Melissa S Love
- Calibr, a division of The Scripps Research Institute , La Jolla, CA, USA
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institute , La Jolla, CA, USA
| |
Collapse
|
20
|
Swale C, Bougdour A, Gnahoui-David A, Tottey J, Georgeault S, Laurent F, Palencia A, Hakimi MA. Metal-captured inhibition of pre-mRNA processing activity by CPSF3 controls Cryptosporidium infection. Sci Transl Med 2020; 11:11/517/eaax7161. [PMID: 31694928 DOI: 10.1126/scitranslmed.aax7161] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022]
Abstract
Cryptosporidium is an intestinal pathogen that causes severe but self-limiting diarrhea in healthy humans, yet it can turn into a life-threatening, unrelenting infection in immunocompromised patients and young children. Severe diarrhea is recognized as the leading cause of mortality for children below 5 years of age in developing countries. The only approved treatment against cryptosporidiosis, nitazoxanide, has limited efficacy in the most vulnerable patient populations, including malnourished children, and is ineffective in immunocompromised individuals. Here, we investigate inhibition of the parasitic cleavage and polyadenylation specificity factor 3 (CPSF3) as a strategy to control Cryptosporidium infection. We show that the oxaborole AN3661 selectively blocked Cryptosporidium growth in human HCT-8 cells, and oral treatment with AN3661 reduced intestinal parasite burden in both immunocompromised and neonatal mouse models of infection with greater efficacy than nitazoxanide. Furthermore, we present crystal structures of recombinantly produced Cryptosporidium CPSF3, revealing a mechanism of action whereby the mRNA processing activity of this enzyme is efficiently blocked by the binding of the oxaborole group at the metal-dependent catalytic center. Our data provide insights that may help accelerate the development of next-generation anti-Cryptosporidium therapeutics.
Collapse
Affiliation(s)
- Christopher Swale
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Alexandre Bougdour
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Audrey Gnahoui-David
- INRA, Université François Rabelais de Tours, Centre Val de Loire, UMR1282 ISP, Laboratoire Apicomplexes et Immunité Mucosale, 37380 Nouzilly, France
| | - Julie Tottey
- INRA, Université François Rabelais de Tours, Centre Val de Loire, UMR1282 ISP, Laboratoire Apicomplexes et Immunité Mucosale, 37380 Nouzilly, France
| | - Sonia Georgeault
- Plateforme des Microscopies, Université et CHRU de Tours, 37000 Tours, France
| | - Fabrice Laurent
- INRA, Université François Rabelais de Tours, Centre Val de Loire, UMR1282 ISP, Laboratoire Apicomplexes et Immunité Mucosale, 37380 Nouzilly, France.
| | - Andrés Palencia
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France. .,Institute for Advanced Biosciences (IAB), Structural Biology of Novel Drug Targets in Human Diseases, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France
| | - Mohamed-Ali Hakimi
- Institute for Advanced Biosciences (IAB), Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, 38000 Grenoble, France.
| |
Collapse
|
21
|
Wang B, Castellanos-Gonzalez A, White AC. Novel drug targets for treatment of cryptosporidiosis. Expert Opin Ther Targets 2020; 24:915-922. [PMID: 32552166 DOI: 10.1080/14728222.2020.1785432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction Cryptosporidium species are protozoan parasites that are important causes of diarrheal disease including waterborne outbreaks, childhood diarrhea in resource-poor countries, and diarrhea in compromised hosts worldwide. Recent studies highlight the importance of cryptosporidiosis in childhood diarrhea, malnutrition, and death in resource-poor countries. Despite this, only a single drug, nitazoxanide, has demonstrated efficacy in human cryptosporidiosis and its efficacy is limited in malnourished children and patients with HIV. Areas covered In this review, we highlight work on potential targets for chemotherapy and review progress on drug development. A number of new targets have been identified for chemotherapy and progress has been made at developing drugs for these targets. Targets include parasite kinases, nucleic acid synthesis and processing, proteases, and lipid metabolism. Other groups have performed high-throughput screening to identify potential drugs. Several compounds have advanced to large animal studies. Expert opinion Development of drugs for cryptosporidiosis has been plagued by a lack of success. Barriers have included poor correlations between in vitro activity and clinical success as well as frequent unanticipated adverse effects. Without a clear pathway forward, it is wise to maintain a diverse development pipeline. Drug developers should also realize that success will likely require a sustained, methodical effort.
Collapse
Affiliation(s)
- Beilin Wang
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch , Galveston, TX, USA
| | | | - A Clinton White
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch , Galveston, TX, USA
| |
Collapse
|
22
|
Arnold SLM, Choi R, Hulverson MA, Whitman GR, Mccloskey MC, Dorr CS, Vidadala RSR, Khatod M, Morada M, Barrett LK, Maly DJ, Yarlett N, Van Voorhis WC. P-Glycoprotein-Mediated Efflux Reduces the In Vivo Efficacy of a Therapeutic Targeting the Gastrointestinal Parasite Cryptosporidium. J Infect Dis 2020; 220:1188-1198. [PMID: 31180118 PMCID: PMC6736360 DOI: 10.1093/infdis/jiz269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/17/2019] [Indexed: 12/04/2022] Open
Abstract
Recent studies have illustrated the burden Cryptosporidium infection places on the lives of malnourished children and immunocompromised individuals. Treatment options remain limited, and efforts to develop a new therapeutic are currently underway. However, there are unresolved questions about the ideal pharmacokinetic characteristics of new anti-Cryptosporidium therapeutics. Specifically, should drug developers optimize therapeutics and formulations to increase drug exposure in the gastrointestinal lumen, enterocytes, or systemic circulation? Furthermore, how should researchers interpret data suggesting their therapeutic is a drug efflux transporter substrate? In vivo drug transporter–mediated alterations in efficacy are well recognized in multiple disease areas, but the impact of intestinal transporters on therapeutic efficacy against enteric diseases has not been established. Using multiple in vitro models and a mouse model of Cryptosporidium infection, we characterized the effect of P-glycoprotein efflux on bumped kinase inhibitor pharmacokinetics and efficacy. Our results demonstrated P-glycoprotein decreases bumped kinase inhibitor enterocyte exposure, resulting in reduced in vivo efficacy against Cryptosporidium. Furthermore, a hollow fiber model of Cryptosporidium infection replicated the in vivo impact of P-glycoprotein on anti-Cryptosporidium efficacy. In conclusion, when optimizing drug candidates targeting the gastrointestinal epithelium or gastrointestinal epithelial infections, drug developers should consider the adverse impact of active efflux transporters on efficacy.
Collapse
Affiliation(s)
- Samuel L M Arnold
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | - Ryan Choi
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | - Matthew A Hulverson
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | - Grant R Whitman
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | - Molly C Mccloskey
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | - Carlie S Dorr
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | | | | | | | - Lynn K Barrett
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle.,Department of Biochemistry, University of Washington, Seattle
| | | | - Wesley C Van Voorhis
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.,Center for Emerging and Reemerging Infectious Disease, University of Washington, Seattle
| |
Collapse
|
23
|
Jumani RS, Spector JM, Izadnegahdar R, Kelly P, Diagana TT, Manjunatha UH. Innovations in Addressing Pediatric Diarrhea in Low Resource Settings. ACS Infect Dis 2020; 6:14-24. [PMID: 31612701 DOI: 10.1021/acsinfecdis.9b00315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Diarrhea has long been recognized as an important cause of mortality during childhood. In parallel with ensuring access to proven care practices is the imperative to apply modern advances in medicine, science, and technology to accelerate progress against diarrheal disease, particularly in developing countries where the burden of avoidable harm is the greatest. In order to highlight achievements and identify outstanding areas of need, we reviewed the landscape of recent innovations that have significance for the study and clinical management of pediatric diarrhea in low resource settings.
Collapse
Affiliation(s)
- Rajiv S. Jumani
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Jonathan M. Spector
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Rasa Izadnegahdar
- Bill and Melinda Gates Foundation, 440 5th Ave N, Seattle, Washington 98109, United States
| | - Paul Kelly
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, Turner Street, London E1 2AD, United Kingdom
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Nationalist Road, Lusaka, Zambia
| | - Thierry T. Diagana
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Ujjini H. Manjunatha
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| |
Collapse
|
24
|
Tosini F, Ludovisi A, Tonanzi D, Amati M, Cherchi S, Pozio E, Gómez-Morales MA. Delivery of SA35 and SA40 peptides in mice enhances humoral and cellular immune responses and confers protection against Cryptosporidium parvum infection. Parasit Vectors 2019; 12:233. [PMID: 31092283 PMCID: PMC6518611 DOI: 10.1186/s13071-019-3486-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/06/2019] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Cryptosporidium parvum is a major cause of diarrhea in children and ruminants at the earliest stages of life. Maternal antibodies represent the main shield of neonate mammals for most of the infections. Two recombinant antigens (SA35 and SA40), portions of two C. parvum proteins, were tested for their ability to induce immune responses in adult mice and for protection on neonate BALB/c mice born from females immunised by mucosal delivery of both peptides. METHODS Adult BALB/c mice were intraperitoneally immunised with SA35 and SA40, separately or mixed, and their immune response was characterised. Furthermore, BALB/c pregnant mice were immunised by mucosal delivery with an SA35/40 mix, before and during pregnancy. Soon after birth, their offspring were infected with two doses (1 × 105 and 5 × 103) of C. parvum oocysts and the parasitic burden was determined at 5 and 9 days post-infection. RESULTS Intraperitoneal immunisation with SA35 and SA40 induced specific IgG and IgG1 in serum, specific IgA in the intestinal mucosa, increase of CD3+/CD4+ and CD30+ cells in splenocytes, which produced IFN-γ. Neonates born from immunised mice and infected with 1 × 105 oocysts showed a significant reduction of oocysts and intestinal forms (23 and 42%, respectively). A reduction of all parasitic forms (96%; P < 0.05) was observed when neonates were infected with 5 × 103 oocysts. CONCLUSIONS SA35 and SA40 peptides induce specific humoral and cell-mediated immune responses to C. parvum in adult mice. Moreover, mucosal administration of the SA35/40 mix in pregnant mice reduces C. parvum burden in their litters.
Collapse
Affiliation(s)
- Fabio Tosini
- European Union Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Ludovisi
- European Union Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Tonanzi
- European Union Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Amati
- European Union Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Cherchi
- European Union Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy
| | - Edoardo Pozio
- European Union Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
25
|
Jumani RS, Hasan MM, Stebbins EE, Donnelly L, Miller P, Klopfer C, Bessoff K, Teixeira JE, Love MS, McNamara CW, Huston CD. A suite of phenotypic assays to ensure pipeline diversity when prioritizing drug-like Cryptosporidium growth inhibitors. Nat Commun 2019; 10:1862. [PMID: 31015448 PMCID: PMC6478823 DOI: 10.1038/s41467-019-09880-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/01/2019] [Indexed: 01/07/2023] Open
Abstract
Cryptosporidiosis is a leading cause of life-threatening diarrhea in children, and the only currently approved drug is ineffective in malnourished children and immunocompromised people. Large-scale phenotypic screens are ongoing to identify anticryptosporidial compounds, but optimal approaches to prioritize inhibitors and establish a mechanistically diverse drug development pipeline are unknown. Here, we present a panel of medium-throughput mode of action assays that enable testing of compounds in several stages of the Cryptosporidium life cycle. Phenotypic profiles are given for thirty-nine anticryptosporidials. Using a clustering algorithm, the compounds sort by phenotypic profile into distinct groups of inhibitors that are either chemical analogs (i.e. same molecular mechanism of action (MMOA)) or known to have similar MMOA. Furthermore, compounds belonging to multiple phenotypic clusters are efficacious in a chronic mouse model of cryptosporidiosis. This suite of phenotypic assays should ensure a drug development pipeline with diverse MMOA without the need to identify underlying mechanisms.
Collapse
Affiliation(s)
- Rajiv S Jumani
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA.,Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT, 05405, USA.,Novartis Institute for Tropical Diseases, Novartis Institutes for BioMedical Research, Emeryville, CA, 94608, USA
| | - Muhammad M Hasan
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA.,Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT, 05405, USA
| | - Erin E Stebbins
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Liam Donnelly
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Peter Miller
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Connor Klopfer
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Kovi Bessoff
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA.,Department of Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305-5101, USA
| | - Jose E Teixeira
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Melissa S Love
- Calibr at The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Case W McNamara
- Calibr at The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Christopher D Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA. .,Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT, 05405, USA. .,Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA.
| |
Collapse
|
26
|
Baragaña B, Forte B, Choi R, Nakazawa Hewitt S, Bueren-Calabuig JA, Pisco JP, Peet C, Dranow DM, Robinson DA, Jansen C, Norcross NR, Vinayak S, Anderson M, Brooks CF, Cooper CA, Damerow S, Delves M, Dowers K, Duffy J, Edwards TE, Hallyburton I, Horst BG, Hulverson MA, Ferguson L, Jiménez-Díaz MB, Jumani RS, Lorimer DD, Love MS, Maher S, Matthews H, McNamara CW, Miller P, O'Neill S, Ojo KK, Osuna-Cabello M, Pinto E, Post J, Riley J, Rottmann M, Sanz LM, Scullion P, Sharma A, Shepherd SM, Shishikura Y, Simeons FRC, Stebbins EE, Stojanovski L, Straschil U, Tamaki FK, Tamjar J, Torrie LS, Vantaux A, Witkowski B, Wittlin S, Yogavel M, Zuccotto F, Angulo-Barturen I, Sinden R, Baum J, Gamo FJ, Mäser P, Kyle DE, Winzeler EA, Myler PJ, Wyatt PG, Floyd D, Matthews D, Sharma A, Striepen B, Huston CD, Gray DW, Fairlamb AH, Pisliakov AV, Walpole C, Read KD, Van Voorhis WC, Gilbert IH. Lysyl-tRNA synthetase as a drug target in malaria and cryptosporidiosis. Proc Natl Acad Sci U S A 2019; 116:7015-7020. [PMID: 30894487 PMCID: PMC6452685 DOI: 10.1073/pnas.1814685116] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Malaria and cryptosporidiosis, caused by apicomplexan parasites, remain major drivers of global child mortality. New drugs for the treatment of malaria and cryptosporidiosis, in particular, are of high priority; however, there are few chemically validated targets. The natural product cladosporin is active against blood- and liver-stage Plasmodium falciparum and Cryptosporidium parvum in cell-culture studies. Target deconvolution in P. falciparum has shown that cladosporin inhibits lysyl-tRNA synthetase (PfKRS1). Here, we report the identification of a series of selective inhibitors of apicomplexan KRSs. Following a biochemical screen, a small-molecule hit was identified and then optimized by using a structure-based approach, supported by structures of both PfKRS1 and C. parvum KRS (CpKRS). In vivo proof of concept was established in an SCID mouse model of malaria, after oral administration (ED90 = 1.5 mg/kg, once a day for 4 d). Furthermore, we successfully identified an opportunity for pathogen hopping based on the structural homology between PfKRS1 and CpKRS. This series of compounds inhibit CpKRS and C. parvum and Cryptosporidium hominis in culture, and our lead compound shows oral efficacy in two cryptosporidiosis mouse models. X-ray crystallography and molecular dynamics simulations have provided a model to rationalize the selectivity of our compounds for PfKRS1 and CpKRS vs. (human) HsKRS. Our work validates apicomplexan KRSs as promising targets for the development of drugs for malaria and cryptosporidiosis.
Collapse
Affiliation(s)
- Beatriz Baragaña
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Barbara Forte
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Ryan Choi
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Stephen Nakazawa Hewitt
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Juan A Bueren-Calabuig
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - João Pedro Pisco
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Caroline Peet
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - David M Dranow
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Beryllium Discovery Corp., Bainbridge Island, WA 98110
| | - David A Robinson
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Chimed Jansen
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Neil R Norcross
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Sumiti Vinayak
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602
| | - Mark Anderson
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Carrie F Brooks
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602
| | - Caitlin A Cooper
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602
| | - Sebastian Damerow
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Michael Delves
- Department of Life Sciences, Imperial College, South Kensington, SW7 2AZ London, United Kingdom
| | - Karen Dowers
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - James Duffy
- Medicines for Malaria Venture, 1215 Geneva 15, Switzerland
| | - Thomas E Edwards
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Beryllium Discovery Corp., Bainbridge Island, WA 98110
| | - Irene Hallyburton
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Benjamin G Horst
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Matthew A Hulverson
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Liam Ferguson
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | | | - Rajiv S Jumani
- Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Donald D Lorimer
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Beryllium Discovery Corp., Bainbridge Island, WA 98110
| | - Melissa S Love
- Biology Department, Calibr at Scripps Research, La Jolla, CA 92037
| | - Steven Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602
| | - Holly Matthews
- Department of Life Sciences, Imperial College, South Kensington, SW7 2AZ London, United Kingdom
| | - Case W McNamara
- Biology Department, Calibr at Scripps Research, La Jolla, CA 92037
| | - Peter Miller
- Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Sandra O'Neill
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Kayode K Ojo
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Maria Osuna-Cabello
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Erika Pinto
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - John Post
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Jennifer Riley
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Matthias Rottmann
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
- Universität Basel, CH-4003 Basel, Switzerland
| | - Laura M Sanz
- Diseases of the Developing World, Global Health, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain
| | - Paul Scullion
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Arvind Sharma
- Structural Parasitology Group, International Centre for Genetic Engineering and Biotechnology, 110067 New Delhi, India
| | - Sharon M Shepherd
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Yoko Shishikura
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Frederick R C Simeons
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Erin E Stebbins
- Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Laste Stojanovski
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Ursula Straschil
- Department of Life Sciences, Imperial College, South Kensington, SW7 2AZ London, United Kingdom
| | - Fabio K Tamaki
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Jevgenia Tamjar
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Leah S Torrie
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 12 201 Phnom Penh, Cambodia
| | - Benoît Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 12 201 Phnom Penh, Cambodia
| | - Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
- Universität Basel, CH-4003 Basel, Switzerland
| | - Manickam Yogavel
- Structural Parasitology Group, International Centre for Genetic Engineering and Biotechnology, 110067 New Delhi, India
| | - Fabio Zuccotto
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | | | - Robert Sinden
- Department of Life Sciences, Imperial College, South Kensington, SW7 2AZ London, United Kingdom
| | - Jake Baum
- Department of Life Sciences, Imperial College, South Kensington, SW7 2AZ London, United Kingdom
| | - Francisco-Javier Gamo
- Diseases of the Developing World, Global Health, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
- Universität Basel, CH-4003 Basel, Switzerland
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602
| | - Elizabeth A Winzeler
- Skaggs School of Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Peter J Myler
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109
- Department of Global Health, University of Washington, Seattle, WA 98195
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195
| | - Paul G Wyatt
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - David Floyd
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - David Matthews
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Amit Sharma
- Structural Parasitology Group, International Centre for Genetic Engineering and Biotechnology, 110067 New Delhi, India
| | - Boris Striepen
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - David W Gray
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Alan H Fairlamb
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Andrei V Pisliakov
- Computational Biology, School of Life Sciences, University of Dundee, DD1 5EH Dundee, United Kingdom
- Physics, School of Science and Engineering, University of Dundee, Dundee DD1 4HN, United Kingdom
| | - Chris Walpole
- Structural Genomics Consortium, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Kevin D Read
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom
| | - Wesley C Van Voorhis
- Seattle Structural Genomics Center for Infectious Disease, Seattle, WA 98109
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109
| | - Ian H Gilbert
- Wellcome Centre for Anti-Infectives Research, Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, DD1 5EH Dundee, United Kingdom;
| |
Collapse
|
27
|
Abstract
The intestinal apicomplexan parasite
Cryptosporidium is a major cause of diarrheal disease in humans worldwide. However, treatment options are severely limited. The search for novel interventions is imperative, yet there are several challenges to drug development, including intractability of the parasite and limited technical tools to study it. This review addresses recent, exciting breakthroughs in this field, including novel cell culture models, strategies for genetic manipulation, transcriptomics, and promising new drug candidates. These advances will stimulate the ongoing quest to understand
Cryptosporidium and the pathogenesis of cryptosporidiosis and to develop new approaches to combat this disease.
Collapse
Affiliation(s)
- Seema Bhalchandra
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts, 02111, USA
| | - Daviel Cardenas
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts, 02111, USA
| | - Honorine D Ward
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts, 02111, USA.,Medicine, Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| |
Collapse
|
28
|
De Rycker M, Baragaña B, Duce SL, Gilbert IH. Challenges and recent progress in drug discovery for tropical diseases. Nature 2018; 559:498-506. [PMID: 30046073 PMCID: PMC6129172 DOI: 10.1038/s41586-018-0327-4] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
Infectious tropical diseases have a huge effect in terms of mortality and morbidity, and impose a heavy economic burden on affected countries. These diseases predominantly affect the world's poorest people. Currently available drugs are inadequate for the majority of these diseases, and there is an urgent need for new treatments. This Review discusses some of the challenges involved in developing new drugs to treat these diseases and highlights recent progress. While there have been notable successes, there is still a long way to go.
Collapse
Affiliation(s)
- Manu De Rycker
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Beatriz Baragaña
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Suzanne L Duce
- Medicines Monitoring Unit (MEMO), Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Ian H Gilbert
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
29
|
|
30
|
Chao AT, Lee BH, Wan KF, Selva J, Zou B, Gedeck P, Beer DJ, Diagana TT, Bonamy GMC, Manjunatha UH. Development of a Cytopathic Effect-Based Phenotypic Screening Assay against Cryptosporidium. ACS Infect Dis 2018; 4:635-645. [PMID: 29341586 DOI: 10.1021/acsinfecdis.7b00247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cryptosporidiosis is a diarrheal disease predominantly caused by Cryptosporidium parvum ( Cp) and Cryptosporidium hominis ( Ch), apicomplexan parasites which infect the intestinal epithelial cells of their human hosts. The only approved drug for cryptosporidiosis is nitazoxanide, which shows limited efficacy in immunocompromised children, the most vulnerable patient population. Thus, new therapeutics and in vitro infection models are urgently needed to address the current unmet medical need. Toward this aim, we have developed novel cytopathic effect (CPE)-based Cp and Ch assays in human colonic tumor (HCT-8) cells and compared them to traditional imaging formats. Further model validation was achieved through screening a collection of FDA-approved drugs and confirming many previously known anti- Cryptosporidium hits as well as identifying a few novel candidates. Collectively, our data reveals this model to be a simple, functional, and homogeneous gain of signal format amenable to high throughput screening, opening new avenues for the discovery of novel anticryptosporidials.
Collapse
Affiliation(s)
- Alexander T. Chao
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Boon Heng Lee
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
| | - Kah Fei Wan
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
| | - Jeremy Selva
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
| | - Bin Zou
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
| | - Peter Gedeck
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
| | - David John Beer
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
| | - Thierry T. Diagana
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Ghislain M. C. Bonamy
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Ujjini H. Manjunatha
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01 Chromos, Singapore, 138670, Singapore
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, California 94608, United States
| |
Collapse
|
31
|
Stebbins E, Jumani RS, Klopfer C, Barlow J, Miller P, Campbell MA, Meyers MJ, Griggs DW, Huston CD. Clinical and microbiologic efficacy of the piperazine-based drug lead MMV665917 in the dairy calf cryptosporidiosis model. PLoS Negl Trop Dis 2018; 12:e0006183. [PMID: 29309415 PMCID: PMC5774826 DOI: 10.1371/journal.pntd.0006183] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/19/2018] [Accepted: 12/20/2017] [Indexed: 02/08/2023] Open
Abstract
Cryptosporidiosis causes life-threatening diarrhea in infants, but the best available treatment is only modestly efficacious. Rodents infected with relevant Cryptosporidium species do not develop diarrhea, which complicates drug development. Cryptosporidium parvum infection of dairy calves, however, causes an illness like that seen in infants. Here, the clinical and microbiologic anti-Cryptosporidium efficacy of the piperazine-based compound MMV665917 was demonstrated in neonatal calves. Oral administration of MMV665917 (22 mg/kg once daily) was begun two days after the onset of severe diarrhea and continued for seven days. Treatment resulted in prompt resolution of diarrhea, and reduced total fecal oocyst shedding by ~94%. MMV665917 was useful for treatment, rather than just prophylaxis, since it was safe and effective when administered well after the onset of diarrhea. Furthermore, even though all animals received intensive supportive care, there was a strong trend towards improved secondary health outcomes, including general health, appetite, and dehydration measures amongst treated animals. These data establish MMV665917 as an outstanding lead compound for Cryptosporidium drug development.
Collapse
Affiliation(s)
- Erin Stebbins
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Rajiv S. Jumani
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
- Cell, Molecular and Biomedical Sciences graduate program, University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Connor Klopfer
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | - John Barlow
- Department of Animal and Veterinary Sciences, University of Vermont College of Agriculture and Life Sciences, Burlington, Vermont
| | - Peter Miller
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | | | | | | | - Christopher D. Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
- Cell, Molecular and Biomedical Sciences graduate program, University of Vermont Larner College of Medicine, Burlington, Vermont
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont
| |
Collapse
|
32
|
Arnold SLM, Choi R, Hulverson MA, Schaefer DA, Vinayak S, Vidadala RSR, McCloskey MC, Whitman GR, Huang W, Barrett LK, Ojo KK, Fan E, Maly DJ, Riggs MW, Striepen B, Van Voorhis WC. Necessity of Bumped Kinase Inhibitor Gastrointestinal Exposure in Treating Cryptosporidium Infection. J Infect Dis 2017; 216:55-63. [PMID: 28541457 PMCID: PMC5853285 DOI: 10.1093/infdis/jix247] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/22/2017] [Indexed: 01/16/2023] Open
Abstract
There is a substantial need for novel therapeutics to combat the widespread impact caused by Crytosporidium infection. However, there is a lack of knowledge as to which drug pharmacokinetic (PK) characteristics are key to generate an in vivo response, specifically whether systemic drug exposure is crucial for in vivo efficacy. To identify which PK properties are correlated with in vivo efficacy, we generated physiologically based PK models to simulate systemic and gastrointestinal drug concentrations for a series of bumped kinase inhibitors (BKIs) that have nearly identical in vitro potency against Cryptosporidium but display divergent PK properties. When BKI concentrations were used to predict in vivo efficacy with a neonatal model of Cryptosporidium infection, these concentrations in the large intestine were the sole predictors of the observed in vivo efficacy. The significance of large intestinal BKI exposure for predicting in vivo efficacy was further supported with an adult mouse model of Cryptosporidium infection. This study suggests that drug exposure in the large intestine is essential for generating a superior in vivo response, and that physiologically based PK models can assist in the prioritization of leading preclinical drug candidates for in vivo testing.
Collapse
Affiliation(s)
- Samuel L M Arnold
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | - Ryan Choi
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | - Matthew A Hulverson
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | - Deborah A Schaefer
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson
| | | | | | - Molly C McCloskey
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | - Grant R Whitman
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | | | - Lynn K Barrett
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | - Kayode K Ojo
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| | - Erkang Fan
- Department of Biochemistry, Biomolecular Structure Center, University of Washington, Seattle
| | | | - Michael W Riggs
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson
| | - Boris Striepen
- Center for Tropical and Emerging Global Diseases
- Department of Cellular Biology, University of Georgia, Athens
| | - Wesley C Van Voorhis
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease
| |
Collapse
|
33
|
Manjunatha UH, Vinayak S, Zambriski JA, Chao AT, Sy T, Noble CG, Bonamy GMC, Kondreddi RR, Zou B, Gedeck P, Brooks CF, Herbert GT, Sateriale A, Tandel J, Noh S, Lakshminarayana SB, Lim SH, Goodman LB, Bodenreider C, Feng G, Zhang L, Blasco F, Wagner J, Leong FJ, Striepen B, Diagana TT. A Cryptosporidium PI(4)K inhibitor is a drug candidate for cryptosporidiosis. Nature 2017; 546:376-380. [PMID: 28562588 PMCID: PMC5473467 DOI: 10.1038/nature22337] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/31/2017] [Indexed: 01/02/2023]
Abstract
Diarrhoeal disease is responsible for 8.6% of global child mortality. Recent epidemiological studies found the protozoan parasite Cryptosporidium to be a leading cause of paediatric diarrhoea, with particularly grave impact on infants and immunocompromised individuals. There is neither a vaccine nor an effective treatment. Here we establish a drug discovery process built on scalable phenotypic assays and mouse models that take advantage of transgenic parasites. Screening a library of compounds with anti-parasitic activity, we identify pyrazolopyridines as inhibitors of Cryptosporidium parvum and Cryptosporidium hominis. Oral treatment with the pyrazolopyridine KDU731 results in a potent reduction in intestinal infection of immunocompromised mice. Treatment also leads to rapid resolution of diarrhoea and dehydration in neonatal calves, a clinical model of cryptosporidiosis that closely resembles human infection. Our results suggest that the Cryptosporidium lipid kinase PI(4)K (phosphatidylinositol-4-OH kinase) is a target for pyrazolopyridines and that KDU731 warrants further preclinical evaluation as a drug candidate for the treatment of cryptosporidiosis.
Collapse
Affiliation(s)
- Ujjini H. Manjunatha
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Sumiti Vinayak
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602 Georgia USA
| | - Jennifer A. Zambriski
- Washington State University, College of Veterinary Medicine, Paul G. Allen School for Global Animal Health, Pullman, 99164 Washington USA
| | - Alexander T. Chao
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Tracy Sy
- Washington State University, College of Veterinary Medicine, Paul G. Allen School for Global Animal Health, Pullman, 99164 Washington USA
| | - Christian G. Noble
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Ghislain M. C. Bonamy
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Ravinder R. Kondreddi
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Bin Zou
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Peter Gedeck
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Carrie F. Brooks
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602 Georgia USA
| | - Gillian T. Herbert
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602 Georgia USA
| | - Adam Sateriale
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602 Georgia USA
| | - Jayesh Tandel
- Department of Cellular Biology, University of Georgia, Athens, 30602 Georgia USA
| | - Susan Noh
- Washington State University, College of Veterinary Medicine, Paul G. Allen School for Global Animal Health, Pullman, 99164 Washington USA
- USDA-Agricultural Research Service, Animal Disease Research Unit, Washington State University, Pullman, 99164 Washington USA
- Department of Veterinary Microbiology and Pathology, Washington State University, Washington Animal Disease Diagnostic Laboratory, Pullman, Washington USA
| | - Suresh B. Lakshminarayana
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Siau H. Lim
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Laura B. Goodman
- Department of Population Medicine and Diagnostic Sciences, Cornell University, College of Veterinary Medicine, Ithaca, 14853 New York USA
| | - Christophe Bodenreider
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Gu Feng
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Lijun Zhang
- China Novartis Institutes for Biomedical Research, Zhangjiang Hi-Tech Park, Pudong, Shanghai, 201203 China
| | - Francesca Blasco
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Juergen Wagner
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - F. Joel Leong
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| | - Boris Striepen
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602 Georgia USA
- Department of Cellular Biology, University of Georgia, Athens, 30602 Georgia USA
| | - Thierry T. Diagana
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, 05-01 Chromos, Singapore, 138670 Singapore
| |
Collapse
|
34
|
Debnath A, McKerrow JH. Editorial: Drug Development for Parasite-Induced Diarrheal Diseases. Front Microbiol 2017; 8:577. [PMID: 28421063 PMCID: PMC5379777 DOI: 10.3389/fmicb.2017.00577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/21/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Anjan Debnath
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - James H McKerrow
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of CaliforniaSan Diego, La Jolla, CA, USA
| |
Collapse
|