1
|
Roig-Sanchis J, Bosch-Nicolau P, Silgado A, Salvador F, Sánchez-Montalvá A, Aznar M, Oliveira I, Espinosa-Pereiro J, Serre-Delcor N, Pou D, Martínez-Campreciós J, Sulleiro E, Molina I. Long-term follow-up of individuals with Chagas disease treated with posaconazole and benznidazole in a non-endemic region: the CHAGASAZOL cohort. Clin Microbiol Infect 2025:S1198-743X(25)00137-5. [PMID: 40157424 DOI: 10.1016/j.cmi.2025.03.018] [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: 12/11/2024] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVES The CHAGASAZOL trial compared posaconazole and benznidazole for treating chronic Chagas disease. Posaconazole showed poor short-term efficacy by means of real-time polymearse chain reaction (qPCR) compared with benznidazole, but few studies have reported long-term follow-up using this tool. The aim of this study was to provide a more comprehensive analysis of the CHAGASAZOL cohort through 11 years of follow-up. METHODS This is a prospective observational cohort of individuals who were included in the CHAGASAZOL trial. Data were censored as of 31 December 2023. Subjects initially treated with posaconazole with a positive qPCR were offered re-treatment with benznidazole. All patients underwent clinical and electrocardiographic evaluations as well as a qPCR at a 6-month or 1-year interval. The primary objective was parasitological failure, defined as any positive qPCR in peripheral blood at any time during follow-up. RESULTS Seventy-two participants were enrolled (median follow-up: 71 months, range 1-147 months). At baseline, 59 (82%) were classified as indeterminate forms, 9 (12%) as cardiac, 2 (3%) as digestive, and 2 (3%) as mixed forms. Forty-eight participants received posaconazole, 45 completing at least 1 follow-up visit. Up to 43 of 45 (95%) presented a positive qPCR, and of them, 35 accepted to be retreated with benznidazole. Considering those treated with benznidazole (either initially or as a re-treatment), only 3 of 51 (6%) showed a positive qPCR. Four (5.5%) participants showed cardiac progression after 3-10 years of follow-up, with an incident rate of 0.94 events per 100 person-years. Two of them had received the complete benznidazole treatment, 1 was partially treated (17 days) and 1 was only treated with posaconazole before clinical progression. DISCUSSION Even if benznidazole showed parasitological efficacy, lifelong follow-up should be offered to individuals living with Chagas disease, as both parasitological failure and clinical progression can occur many years after diagnosis and treatment.
Collapse
Affiliation(s)
- Joan Roig-Sanchis
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
| | - Pau Bosch-Nicolau
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| | - Aroa Silgado
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; Microbiology Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
| | - Fernando Salvador
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Adrián Sánchez-Montalvá
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marisa Aznar
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Inés Oliveira
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Espinosa-Pereiro
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Serre-Delcor
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Diana Pou
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Martínez-Campreciós
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Sulleiro
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; Microbiology Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
| | - Israel Molina
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Ernst L, Macedo GC, McCall LI. System-based insights into parasitological and clinical treatment failure in Chagas disease. mSystems 2025; 10:e0003824. [PMID: 39772644 PMCID: PMC11834445 DOI: 10.1128/msystems.00038-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Infectious disease treatment success requires symptom resolution (clinical treatment success), which often but not always involves pathogen clearance. Both of these treatment goals face disease-specific and general challenges. In this review, we summarize the current state of knowledge in mechanisms of clinical and parasitological treatment failure in the context of Chagas disease, a neglected tropical disease causing cardiac and gastrointestinal symptoms. Parasite drug resistance and persistence, drug pharmacokinetics and dynamics, as well as persistently altered host immune responses and tissue damage are the most common reasons for Chagas disease treatment failure. We discuss the therapeutics that failed before regulatory approval, limitations of current therapeutic options and new treatment strategies to overcome persistent parasites, inflammatory responses, and metabolic alterations. Large-scale omics analyses were critical in generating these insights and will continue to play a prominent role in addressing the challenges still facing Chagas disease drug treatment.
Collapse
Affiliation(s)
- Luis Ernst
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| | - Giovana C. Macedo
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
3
|
Garg NJ. An Update on Vaccines Against Trypanosoma cruzi and Chagas Disease. Pathogens 2025; 14:124. [PMID: 40005501 PMCID: PMC11857938 DOI: 10.3390/pathogens14020124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Chagas disease (CD) is a global health concern, with no existing therapies to prophylactically treat adults traveling to endemic countries or those who may already be infected with Trypanosoma cruzi. The economic burden of Chagas cardiomyopathy and heart failure, due to healthcare costs and lost productivity from premature deaths, provides a strong rationale for investment in the development of immune therapies against CD. Vaccine efficacy is proposed to depend heavily on the induction of a robust Th1 response for the clearance of intracellular pathogens like T. cruzi. In this review, updated information on the efforts for vaccine development against CD is provided.
Collapse
Affiliation(s)
- Nisha J. Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch (UTMB), Galveston, TX 77555-1070, USA;
- Institute for Human Infections and Immunity, University of Texas Medical Branch (UTMB), Galveston, TX 77555-1070, USA
| |
Collapse
|
4
|
Eadsforth TC, Torrie LS, Rowland P, Edgar EV, MacLean LM, Paterson C, Robinson DA, Shepherd SM, Thomas J, Thomas MG, Gray DW, Postis VLG, De Rycker M. Pharmacological and structural understanding of the Trypanosoma cruzi proteasome provides key insights for developing site-specific inhibitors. J Biol Chem 2025; 301:108049. [PMID: 39638245 PMCID: PMC11748689 DOI: 10.1016/j.jbc.2024.108049] [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: 05/22/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The proteasome is considered an excellent drug target for many infectious diseases as well as cancer. Challenges with robust and safe supply of proteasomes from infectious agents, lack of structural information, and complex pharmacology due to multiple active sites have hampered progress in the infectious disease space. We recombinantly expressed the proteasome of the protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease, and demonstrate pharmacological equivalence to the native T. cruzi proteasome. Active-site mutant recombinant proteasomes reveal substrate promiscuity for WT proteasomes, with important implications for assessing pharmacological responses of active-site selective inhibitors. Using these mutant proteasomes, we show that some selective parasite proteasome inhibitors only partially inhibit the chymotrypsin-like activity, including a newly developed 5-(phenoxymethyl)furan-2-carboxamide-based proteasome inhibitor. In spite of partial inhibition, these compounds remain potent inhibitors of intracellular T. cruzi growth. Drug-resistant mutants provide further insights in drug mode-of-inhibition. We also present the high-resolution CryoEM structures of both native and recombinantly-expressed T. cruzi proteasomes which reveal pharmacologically relevant differences in the ligand-binding site compared to the related Leishmania proteasome. Furthermore, we show that the trypanosomatid β4/β5 selectivity pocket is not present in the proteasome structures of other protozoan parasites. This work highlights the need, and provides approaches, to precisely assess proteasome substrate selectivity and pharmacology. It enables structure-guided drug discovery for this promising Chagas disease drug target, provides a new chemical starting point for drug discovery, and paves the road for development of robust proteasome drug discovery programmes for other eukaryotic infectious diseases.
Collapse
Affiliation(s)
- Thomas C Eadsforth
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Leah S Torrie
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | - Lorna M MacLean
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Christy Paterson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - David A Robinson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Sharon M Shepherd
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - John Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Michael G Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - David W Gray
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Vincent L G Postis
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Manu De Rycker
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK.
| |
Collapse
|
5
|
Khan AA, Taylor MC, Fortes Francisco A, Jayawardhana S, Atherton RL, Olmo F, Lewis MD, Kelly JM. Animal models for exploring Chagas disease pathogenesis and supporting drug discovery. Clin Microbiol Rev 2024; 37:e0015523. [PMID: 39545730 PMCID: PMC11629624 DOI: 10.1128/cmr.00155-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
SUMMARYInfections with the parasitic protozoan Trypanosoma cruzi cause Chagas disease, which results in serious cardiac and/or digestive pathology in 30%-40% of individuals. However, symptomatic disease can take decades to become apparent, and there is a broad spectrum of possible outcomes. The complex and long-term nature of this infection places a major constraint on the scope for experimental studies in humans. Accordingly, predictive animal models have been a mainstay of Chagas disease research. The resulting data have made major contributions to our understanding of parasite biology, immune responses, and disease pathogenesis and have provided a platform that informs and facilitates the global drug discovery effort. Here, we provide an overview of available animal models and illustrate how they have had a key impact across the field.
Collapse
Affiliation(s)
- Archie A. Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Richard L. Atherton
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
6
|
Lozano N, Prescilla-Ledezma A, Calabuig E, Trelis M, Arce JMS, López Hontangas JL, de Pablos LM, Gomez-Samblas M, Osuna A. Circulating extracellular vesicles in sera of chronic patients as a method for determining active parasitism in Chagas disease. PLoS Negl Trop Dis 2024; 18:e0012356. [PMID: 39565824 PMCID: PMC11616892 DOI: 10.1371/journal.pntd.0012356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/04/2024] [Accepted: 10/30/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Chagas disease, once restricted mainly to the Americas, Chagas disease has become a global health problem due to migration from endemic to non-endemic areas. In non-endemic regions, transmission is limited to vertical transmission from infected mothers to newborns or through blood and organ donations. A major challenge in the management of the disease lies in the diagnosis of chronic cases, as blood-borne parasites are often absent and antibodies persist for life, complicating the evaluation of treatment. METHODOLOGY AND MAIN FINDINGS This study investigates whether detection of circulating extracellular vesicles (EVs) or their immunocomplexes with host IgGs in the serum of chronic patients with Chagas disease could serve as diagnostic tools and biomarkers of the active presence of the parasite. This method may prove valuable in cases where parasitaemia and other diagnostic tests are inconclusive, especially for assessing treatment efficacy and confirming mother-to-child transmission. Together with exovesicle purification by ultracentrifugation, which is the 'gold standard', an affordable and simplified method for the isolation of EVs or immunocomplexes was tested for use in less well-equipped diagnostic laboratories. EV detection was performed by enzyme-linked immunosorbent assay (ELISA) targeting Trypanosoma cruzi antigens. Positive results were demonstrated in Bolivian patients in Spain, covering asymptomatic and symptomatic cases (cardiac, gastrointestinal or both). The study also examined infected mothers and their newborns. These findings were further confirmed in Panamanian patients with inconclusive diagnostic results. Moreover, host IgG isotypes that formed immunocomplexes with parasite exovsicles were identified, with IgG2 and IgG4 being predominant. CONCLUSIONS Our results confirm the usefulness of circulating EVs and their immunocomplexes as markers of metabolically active T. cruzi in chronic infections without detectable parasitaemia, as well as their efficacy in confirming vertical transmission and in cases of inconclusive diagnostic tests.
Collapse
Affiliation(s)
- Noelia Lozano
- Area of Parasitology, Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Burjassot, Valencia, Spain
- Department of Clinical Microbiology and Parasitology, Hospital Universitario y Politécnico La Fe, Martorell, Valencia, Spain
| | - Alexa Prescilla-Ledezma
- Department of Human Microbiology, Faculty of Medicine, University of Panama, Panama
- Department of Parasitology, Biochemical and Molecular Parasitology Group CTS-183, and Institute of Biotechnology, University of Granada., Granada, Spain
| | - Eva Calabuig
- Infectious Diseases Unit, Internal Medicine Department, Hospital Universitario y Politécnico La Fe, Martorell, Valencia, Spain
- University of Valencia-Health Research Institute La Fe-IIS, Valencia, Spain
| | - Maria Trelis
- Area of Parasitology, Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Burjassot, Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute La Fe-IIS, Valencia, Spain
| | - José Miguel Sahuquillo Arce
- Department of Clinical Microbiology and Parasitology, Hospital Universitario y Politécnico La Fe, Martorell, Valencia, Spain
| | - José Luis López Hontangas
- Department of Clinical Microbiology and Parasitology, Hospital Universitario y Politécnico La Fe, Martorell, Valencia, Spain
| | - Luis Miguel de Pablos
- Department of Parasitology, Biochemical and Molecular Parasitology Group CTS-183, and Institute of Biotechnology, University of Granada., Granada, Spain
| | - Mercedes Gomez-Samblas
- Department of Parasitology, Biochemical and Molecular Parasitology Group CTS-183, and Institute of Biotechnology, University of Granada., Granada, Spain
| | - Antonio Osuna
- Department of Parasitology, Biochemical and Molecular Parasitology Group CTS-183, and Institute of Biotechnology, University of Granada., Granada, Spain
| |
Collapse
|
7
|
Tomasina R, González FC, Cabrera A, Basmadjián Y, Robello C. From Trypomastigotes to Trypomastigotes: Analyzing the One-Way Intracellular Journey of Trypanosoma cruzi by Ultrastructure Expansion Microscopy. Pathogens 2024; 13:866. [PMID: 39452737 PMCID: PMC11510640 DOI: 10.3390/pathogens13100866] [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: 08/11/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
The protozoan parasite Trypanosoma cruzi is the causative agent of Chagas disease, also called American trypanosomiasis. This neglected tropical disease affects millions of individuals across the Americas. To complete its life cycle, T. cruzi parasitizes both vertebrate hosts and its vector, commonly known as the 'kissing bug'. The parasite's survival and proliferation strategies are driven by the diverse environments it encounters. Despite being described by Carlos Chagas in 1909, significant knowledge gaps persist regarding the parasite's various life forms and adaptive capabilities in response to environmental cues. In this study, we employed Ultrastructure Expansion Microscopy to explore the intricate journey of T. cruzi within the host cell. Upon entry into the host cell, trypomastigotes undergo folding, resulting in intermediate forms characterized by a rounded cell body, anterior positioning of basal bodies, and a shortened flagellum. The repositioning of basal bodies and the kinetoplast and the shortening of the flagella mark the culmination of intracellular amastigogenesis. Furthermore, we analyzed intracellular trypomastigogenesis, identifying discrete intermediate forms, including leaf-shaped stages and epimastigote-like forms, which suggests a complex differentiation process. Notably, we did not observe any dividing intracellular epimastigotes, indicating that these may be non-replicative forms within the host cell. Our detailed examination of amastigote cell division revealed semi-closed nuclear mitosis, with mitotic spindle formation independent of basal bodies. This study provides new insights into the morphological and cytoskeletal changes during the intracellular stages of T. cruzi, providing a model for understanding the dynamics of intracellular amastigogenesis and trypomastigogenesis.
Collapse
Affiliation(s)
- Ramiro Tomasina
- Laboratorio de Interacciones Hospedero Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (R.T.); (F.C.G.); (A.C.)
- Unidad Académica de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo 11550, Uruguay;
| | - Fabiana C. González
- Laboratorio de Interacciones Hospedero Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (R.T.); (F.C.G.); (A.C.)
| | - Andrés Cabrera
- Laboratorio de Interacciones Hospedero Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (R.T.); (F.C.G.); (A.C.)
- Unidad Académica de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo 11550, Uruguay;
| | - Yester Basmadjián
- Unidad Académica de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo 11550, Uruguay;
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; (R.T.); (F.C.G.); (A.C.)
- Unidad Académica de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11550, Uruguay
| |
Collapse
|
8
|
Pinazo MJ, Malchiodi E, Ioset JR, Bivona A, Gollob KJ, Dutra WO. Challenges and advancements in the development of vaccines and therapies against Chagas disease. THE LANCET. MICROBE 2024; 5:100972. [PMID: 39303738 DOI: 10.1016/j.lanmic.2024.100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, presents a substantial global health burden, affecting millions of individuals worldwide and posing a continual risk of infection. Despite the high mortality and morbidity rates, effective vaccines to prevent infection by the parasite remain elusive, and the drugs currently available are suboptimal. Understanding the intricate dynamics of parasite-host interactions and the resulting immune responses, which contribute to both protection and pathology, is crucial for the development of effective vaccines and therapies against Chagas disease. In this Series paper, we discuss the challenges associated with discovering and translating prophylactic and therapeutic strategies from the laboratory bench to clinical application. We highlight ongoing efforts in vaccine and new drug development, with a focus on more advanced candidates for vaccines and drugs. We also discuss potential solutions, emphasising the importance of collaborative research efforts, sustained funding, and a comprehensive understanding of host-parasite interactions and immunopathology to advance the development of new vaccines and therapies against Chagas disease.
Collapse
Affiliation(s)
| | - Emilio Malchiodi
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Instituto de Estudios de la Inmunidad Humoral (IDEHU) and Instituto de Microbiologia y Parasitologia Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | | | - Augusto Bivona
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Instituto de Estudios de la Inmunidad Humoral (IDEHU) and Instituto de Microbiologia y Parasitologia Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Kenneth J Gollob
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Belo Horizonte, Brazil
| | - Walderez O Dutra
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Belo Horizonte, Brazil; Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
9
|
Silva TA, Thomas D, Siqueira-Neto JL, Calvet CM. Pirfenidone Prevents Heart Fibrosis during Chronic Chagas Disease Cardiomyopathy. Int J Mol Sci 2024; 25:7302. [PMID: 39000409 PMCID: PMC11242150 DOI: 10.3390/ijms25137302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 07/16/2024] Open
Abstract
Cardiac fibrosis is a severe outcome of Chagas disease (CD), caused by the protozoan Trypanosoma cruzi. Clinical evidence revealed a correlation between fibrosis levels with impaired cardiac performance in CD patients. Therefore, we sought to analyze the effect of inhibitors of TGF-β (pirfenidone), p38-MAPK (losmapimod) and c-Jun (SP600125) on the modulation of collagen deposition in cardiac fibroblasts (CF) and in vivo models of T. cruzi chronic infection. Sirius Red/Fast Green dye was used to quantify both collagen expression and total protein amount, assessing cytotoxicity. The compounds were also used to treat C57/Bl6 mice chronically infected with T. cruzi, Brazil strain. We identified an anti-fibrotic effect in vitro for pirfenidone (TGF-β inhibitor, IC50 114.3 μM), losmapimod (p38 inhibitor, IC50 17.6 μM) and SP600125 (c-Jun inhibitor, IC50 3.9 μM). This effect was independent of CF proliferation since these compounds do not affect T. cruzi-induced host cell multiplication as measured by BrdU incorporation. Assays of chronic infection of mice with T. cruzi have shown a reduction in heart collagen by pirfenidone. These results propose a novel approach to fibrosis therapy in CD, with the prospect of repurposing pirfenidone to prevent the onset of ECM accumulation in the hearts of the patients.
Collapse
Affiliation(s)
- Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Diane Thomas
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.T.); (J.L.S.-N.)
| | - Jair L. Siqueira-Neto
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.T.); (J.L.S.-N.)
| | - Claudia Magalhaes Calvet
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| |
Collapse
|
10
|
Soeiro MDNC, Sales-Junior PA, Pereira VRA, Vannier-Santos MA, Murta SMF, de Sousa AS, Sangenis LHC, Moreno AMH, Boechat N, Branco FSC, Holetz FB, Ávila AR, Pereira MCDS. Drug screening and development cascade for Chagas disease: an update of in vitro and in vivo experimental models. Mem Inst Oswaldo Cruz 2024; 119:e240057. [PMID: 38958341 PMCID: PMC11218046 DOI: 10.1590/0074-02760240057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/03/2024] [Indexed: 07/04/2024] Open
Abstract
Chagas disease is a tropical neglected disease that affects millions of people worldwide, still demanding a more effective and safer therapy, especially in its chronic phase which lacks a treatment that promotes substantial parasitological cure. The technical note of Romanha and collaborators published in 2010 aimed establish a guideline with the set of minimum criteria and decision gates for the development of new agents against Trypanosoma cruzi with the focus on developing new antichagasic drugs. In this sense, the present review aims to update this technical note, bringing the state of the art and new advances on this topic in recent years.
Collapse
Affiliation(s)
| | - Policarpo Ademar Sales-Junior
- Fundação Oswaldo Cruz-Fiocruz, Instituto Aggeu Magalhães, Departamento de Imunologia, Laboratório de Imunopatologia e Biologia Molecular, Recife, PE, Brasil
| | - Valeria Rêgo Alves Pereira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Aggeu Magalhães, Departamento de Imunologia, Laboratório de Imunopatologia e Biologia Molecular, Recife, PE, Brasil
| | - Marcos André Vannier-Santos
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | | | - Andrea Silvestre de Sousa
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em Doença de Chagas, Rio de Janeiro, RJ, Brasil
| | - Luiz Henrique Conde Sangenis
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em Doença de Chagas, Rio de Janeiro, RJ, Brasil
| | - Alejandro Marcel Hasslocher Moreno
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em Doença de Chagas, Rio de Janeiro, RJ, Brasil
| | - Núbia Boechat
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Fármacos - Farmanguinhos, Departamento de Síntese de Fármacos e Bioativos, Laboratório de Síntese de Fármacos, Rio de Janeiro, RJ, Brasil
| | - Frederico Silva Castelo Branco
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Fármacos - Farmanguinhos, Departamento de Síntese de Fármacos e Bioativos, Laboratório de Síntese de Fármacos, Rio de Janeiro, RJ, Brasil
| | - Fabíola Barbieri Holetz
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Regulação da Expressão Gênica, Curitiba, PR, Brasil
| | - Andrea Rodrigues Ávila
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Pesquisa em Apicomplexa, Curitiba, PR, Brasil
| | | |
Collapse
|
11
|
Silveira ACA, Uombe NPI, Velikkakam T, Borges BC, Teixeira TL, de Almeida VF, Torres JDA, Pereira CL, de Souza G, Teixeira SC, Servato JPS, Silva MJB, Mineo TWP, Ribas RM, Mortara RA, da Silveira JF, da Silva CV. The Trypanosoma cruzi pleiotropic protein P21 orchestrates the intracellular retention and in-vivo parasitism control of virulent Y strain parasites. Front Cell Infect Microbiol 2024; 14:1412345. [PMID: 38988814 PMCID: PMC11233463 DOI: 10.3389/fcimb.2024.1412345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
P21 is a protein secreted by all forms of Trypanosoma cruzi (T. cruzi) with recognized biological activities determined in studies using the recombinant form of the protein. In our recent study, we found that the ablation of P21 gene decreased Y strain axenic epimastigotes multiplication and increased intracellular replication of amastigotes in HeLa cells infected with metacyclic trypomastigotes. In the present study, we investigated the effect of P21 in vitro using C2C12 cell lines infected with tissue culture-derived trypomastigotes (TCT) of wild-type and P21 knockout (TcP21-/-) Y strain, and in vivo using an experimental model of T. cruzi infection in BALB/c mice. Our in-vitro results showed a significant decrease in the host cell invasion rate by TcP21-/- parasites as measured by Giemsa staining and cell count in bright light microscope. Quantitative polymerase chain reaction (qPCR) analysis showed that TcP21-/- parasites multiplied intracellularly to a higher extent than the scrambled parasites at 72h post-infection. In addition, we observed a higher egress of TcP21-/- trypomastigotes from C2C12 cells at 144h and 168h post-infection. Mice infected with Y strain TcP21-/- trypomastigotes displayed higher systemic parasitemia, heart tissue parasite burden, and several histopathological alterations in heart tissues compared to control animals infected with scrambled parasites. Therewith, we propose that P21 is important in the host-pathogen interaction during invasion, cell multiplication, and egress, and may be part of the mechanism that controls parasitism and promotes chronic infection without patent systemic parasitemia.
Collapse
Affiliation(s)
| | - Nelsa Paula Inácio Uombe
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Teresiama Velikkakam
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Thaise Lara Teixeira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Cecília Luiza Pereira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Guilherme de Souza
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | | | - Rosineide Marques Ribas
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
12
|
Torchelsen FKVDS, Mazzeti AL, Mosqueira VCF. Drugs in preclinical and early clinical development for the treatment of Chagas´s disease: the current status. Expert Opin Investig Drugs 2024; 33:575-590. [PMID: 38686546 DOI: 10.1080/13543784.2024.2349289] [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: 02/09/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Chagas disease is spreading faster than expected in different countries, and little progress has been reported in the discovery of new drugs to combat Trypanosoma cruzi infection in humans. Recent clinical trials have ended with small hope. The pathophysiology of this neglected disease and the genetic diversity of parasites are exceptionally complex. The only two drugs available to treat patients are far from being safe, and their efficacy in the chronic phase is still unsatisfactory. AREAS COVERED This review offers a comprehensive examination and critical review of data reported in the last 10 years, and it is focused on findings of clinical trials and data acquired in vivo in preclinical studies. EXPERT OPINION The in vivo investigations classically in mice and dog models are also challenging and time-consuming to attest cure for infection. Poorly standardized protocols, availability of diagnosis methods and disease progression markers, the use of different T. cruzi strains with variable benznidazole sensitivities, and animals in different acute and chronic phases of infection contribute to it. More synchronized efforts between research groups in this field are required to put in evidence new promising substances, drug combinations, repurposing strategies, and new pharmaceutical formulations to impact the therapy.
Collapse
Affiliation(s)
- Fernanda Karoline Vieira da Silva Torchelsen
- School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Brazil
- Post-Graduation Program in Pharmaceutical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lia Mazzeti
- Department of Biomedical Sciences and Health, Academic Unit of Passos, University of Minas Gerais State, Passos, Brazil
| | | |
Collapse
|
13
|
Murta SMF, Lemos Santana PA, Jacques Dit Lapierre TJW, Penteado AB, El Hajje M, Navarro Vinha TC, Liarte DB, de Souza ML, Goulart Trossini GH, de Oliveira Rezende Júnior C, de Oliveira RB, Ferreira RS. New drug discovery strategies for the treatment of benznidazole-resistance in Trypanosoma cruzi, the causative agent of Chagas disease. Expert Opin Drug Discov 2024; 19:741-753. [PMID: 38715393 DOI: 10.1080/17460441.2024.2349155] [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: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Benznidazole, the drug of choice for treating Chagas Disease (CD), has significant limitations, such as poor cure efficacy, mainly in the chronic phase of CD, association with side effects, and parasite resistance. Understanding parasite resistance to benznidazole is crucial for developing new drugs to treat CD. AREAS COVERED Here, the authors review the current understanding of the molecular basis of benznidazole resistance. Furthermore, they discuss the state-of-the-art methods and critical outcomes employed to evaluate the efficacy of potential drugs against T. cruzi, aiming to select better compounds likely to succeed in the clinic. Finally, the authors describe the different strategies employed to overcome resistance to benznidazole and find effective new treatments for CD. EXPERT OPINION Resistance to benznidazole is a complex phenomenon that occurs naturally among T. cruzi strains. The combination of compounds that inhibit different metabolic pathways of the parasite is an important strategy for developing a new chemotherapeutic protocol.
Collapse
Affiliation(s)
- Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos - Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Pedro Augusto Lemos Santana
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - André Berndt Penteado
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Marissa El Hajje
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Mariana Laureano de Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
14
|
da Silva CV, Velikkakam T, de Oliveira ECM, Silveira ACA, de Lima Júnior JP, Uombe NPI, da Silva PHR, Borges BC. Cellular dormancy: A widespread phenomenon that perpetuates infectious diseases. J Basic Microbiol 2024; 64:e2300389. [PMID: 38064123 DOI: 10.1002/jobm.202300389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 05/03/2024]
Abstract
Under adverse environmental conditions, microorganisms are able to enter a state of cellular dormancy which consists of cell cycle arrest and interruption of multiplication. This process ensures their perpetuation in the infected host organism and enables the spread of disease. Throughout biological evolution, dormancy allowed microorganisms to persist in a harsh niche until favorable conditions for their reactivation were re-established. Here, we propose to discuss the dormancy of bacteria and protozoa pathogens focusing on the potential mechanisms and components associated with dormancy.
Collapse
Affiliation(s)
- Claudio V da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Teresiama Velikkakam
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Elida C M de Oliveira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Anna C A Silveira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Joed P de Lima Júnior
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Nelsa P I Uombe
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Paulo H R da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Bruna C Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
15
|
Carey SM, O’Neill DM, Conner GB, Sherman J, Rodriguez A, D’Antonio EL. Discovery of Strong 3-Nitro-2-Phenyl- 2H-Chromene Analogues as Antitrypanosomal Agents and Inhibitors of Trypanosoma cruzi Glucokinase. Int J Mol Sci 2024; 25:4319. [PMID: 38673904 PMCID: PMC11050443 DOI: 10.3390/ijms25084319] [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: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chagas disease is one of the world's neglected tropical diseases, caused by the human pathogenic protozoan parasite Trypanosoma cruzi. There is currently a lack of effective and tolerable clinically available therapeutics to treat this life-threatening illness and the discovery of modern alternative options is an urgent matter. T. cruzi glucokinase (TcGlcK) is a potential drug target because its product, d-glucose-6-phosphate, serves as a key metabolite in the pentose phosphate pathway, glycolysis, and gluconeogenesis. In 2019, we identified a novel cluster of TcGlcK inhibitors that also exhibited anti-T. cruzi efficacy called the 3-nitro-2-phenyl-2H-chromene analogues. This was achieved by performing a target-based high-throughput screening (HTS) campaign of 13,040 compounds. The selection criteria were based on first determining which compounds strongly inhibited TcGlcK in a primary screen, followed by establishing on-target confirmed hits from a confirmatory assay. Compounds that exhibited notable in vitro trypanocidal activity over the T. cruzi infective form (trypomastigotes and intracellular amastigotes) co-cultured in NIH-3T3 mammalian host cells, as well as having revealed low NIH-3T3 cytotoxicity, were further considered. Compounds GLK2-003 and GLK2-004 were determined to inhibit TcGlcK quite well with IC50 values of 6.1 µM and 4.8 µM, respectively. Illuminated by these findings, we herein screened a small compound library consisting of thirteen commercially available 3-nitro-2-phenyl-2H-chromene analogues, two of which were GLK2-003 and GLK2-004 (compounds 1 and 9, respectively). Twelve of these compounds had a one-point change from the chemical structure of GLK2-003. The analogues were run through a similar primary screening and confirmatory assay protocol to our previous HTS campaign. Subsequently, three in vitro biological assays were performed where compounds were screened against (a) T. cruzi (Tulahuen strain) infective form co-cultured within NIH-3T3 cells, (b) T. brucei brucei (427 strain) bloodstream form, and (c) NIH-3T3 host cells alone. We report on the TcGlcK inhibitor constant determinations, mode of enzyme inhibition, in vitro antitrypanosomal IC50 determinations, and an assessment of structure-activity relationships. Our results reveal that the 3-nitro-2-phenyl-2H-chromene scaffold holds promise and can be further optimized for both Chagas disease and human African trypanosomiasis early-stage drug discovery research.
Collapse
Affiliation(s)
- Shane M. Carey
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Destiny M. O’Neill
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Garrett B. Conner
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA (A.R.)
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA (A.R.)
| | - Edward L. D’Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| |
Collapse
|
16
|
Silvestrini MMA, Alessio GD, Frias BED, Sales Júnior PA, Araújo MSS, Silvestrini CMA, Brito Alvim de Melo GE, Martins-Filho OA, Teixeira-Carvalho A, Martins HR. New insights into Trypanosoma cruzi genetic diversity, and its influence on parasite biology and clinical outcomes. Front Immunol 2024; 15:1342431. [PMID: 38655255 PMCID: PMC11035809 DOI: 10.3389/fimmu.2024.1342431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, remains a serious public health problem worldwide. The parasite was subdivided into six distinct genetic groups, called "discrete typing units" (DTUs), from TcI to TcVI. Several studies have indicated that the heterogeneity of T. cruzi species directly affects the diversity of clinical manifestations of Chagas disease, control, diagnosis performance, and susceptibility to treatment. Thus, this review aims to describe how T. cruzi genetic diversity influences the biology of the parasite and/or clinical parameters in humans. Regarding the geographic dispersion of T. cruzi, evident differences were observed in the distribution of DTUs in distinct areas. For example, TcII is the main DTU detected in Brazilian patients from the central and southeastern regions, where there are also registers of TcVI as a secondary T. cruzi DTU. An important aspect observed in previous studies is that the genetic variability of T. cruzi can impact parasite infectivity, reproduction, and differentiation in the vectors. It has been proposed that T. cruzi DTU influences the host immune response and affects disease progression. Genetic aspects of the parasite play an important role in determining which host tissues will be infected, thus heavily influencing Chagas disease's pathogenesis. Several teams have investigated the correlation between T. cruzi DTU and the reactivation of Chagas disease. In agreement with these data, it is reasonable to suppose that the immunological condition of the patient, whether or not associated with the reactivation of the T. cruzi infection and the parasite strain, may have an important role in the pathogenesis of Chagas disease. In this context, understanding the genetics of T. cruzi and its biological and clinical implications will provide new knowledge that may contribute to additional strategies in the diagnosis and clinical outcome follow-up of patients with Chagas disease, in addition to the reactivation of immunocompromised patients infected with T. cruzi.
Collapse
Affiliation(s)
| | - Glaucia Diniz Alessio
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Bruna Estefânia Diniz Frias
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Policarpo Ademar Sales Júnior
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Márcio Sobreira Silva Araújo
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Olindo Assis Martins-Filho
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Andréa Teixeira-Carvalho
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Helen Rodrigues Martins
- Department of Pharmacy, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Minas Gerais, Brazil
| |
Collapse
|
17
|
Pinazo MJ, Forsyth C, Losada I, Esteban ET, García-Rodríguez M, Villegas ML, Molina I, Crespillo-Andújar C, Gállego M, Ballart C, Ramirez JC, Aden T, Hoerauf A, Pfarr K, Vaillant M, Marques T, Fernandes J, Blum B, Ribeiro I, Sosa-Estani S, Barreira F, Gascón J. Efficacy and safety of fexinidazole for treatment of chronic indeterminate Chagas disease (FEXI-12): a multicentre, randomised, double-blind, phase 2 trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:395-403. [PMID: 38218194 DOI: 10.1016/s1473-3099(23)00651-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/30/2023] [Accepted: 10/12/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND More than six million people worldwide, particularly in vulnerable communities in Latin America, are infected with Trypanosoma cruzi, the causative agent of Chagas disease. Only a small portion have access to diagnosis and treatment. Both drugs used to treat this chronic, neglected infection, benznidazole and nifurtimox, were developed more than 50 years ago, and adverse drug reactions during treatment pose a major barrier, causing 20% of patients to discontinue therapy. Fexinidazole proved efficacious in an earlier, interrupted clinical trial, but the doses evaluated were not well tolerated. The present study evaluated fexinidazole at lower doses and for shorter treatment durations. METHODS In this randomised, double-blind, phase 2 trial, we included adult patients (18-60 years old) with confirmed T cruzi infection by serology and PCR and without signs of organ involvement. We evaluated three regimens of fexinidazole-600 mg once daily for 10 days (6·0 g total dose), 1200 mg daily for 3 days (3·6 g), and 600 mg daily for 3 days followed by 1200 mg daily for 4 days (6·6 g)-and compared them with a historical placebo control group (n=47). The primary endpoint was sustained negative results by PCR at end of treatment and on each visit up to four months of follow-up. This study is registered with ClinicalTrials.gov, NCT03587766, and EudraCT, 2016-004905-15. FINDINGS Between Oct 16, 2017, and Aug 7, 2018, we enrolled 45 patients (n=15 for each group), of whom 43 completed the study. Eight (19%) of 43 fexinidazole-treated patients reached the primary endpoint, compared with six (13%) of 46 in the historical control group. Mean parasite load decreased sharply following treatment but rebounded beginning 10 weeks after treatment. Five participants had seven grade 3 adverse events: carpal tunnel, sciatica, device infection, pneumonia, staphylococcal infection, and joint and device dislocation. Two participants discontinued treatment due to adverse events unrelated to fexinidazole. INTERPRETATION The fexinidazole regimens in this study had an acceptable safety profile but did not prove effective against T cruzi infection. Development of fexinidazole monotherapy for treating T cruzi infection has been stopped. FUNDING The Drugs for Neglected Diseases initiative.
Collapse
Affiliation(s)
- Maria-Jesus Pinazo
- Barcelona Institute for Global Health, ISGlobal-Hospital Clinic, Universitat de Barcelona, Barcelona, Spain; Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil; CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Spain.
| | - Colin Forsyth
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil
| | - Irene Losada
- Barcelona Institute for Global Health, ISGlobal-Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Elena Trigo Esteban
- Unidad de Patología Importada y Salud Internacional. Hospital Universitario La Paz, Madrid, Spain
| | - Magdalena García-Rodríguez
- Infectious Diseases Service, International Health Unit, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Maria Luz Villegas
- Department of Internal Medicine, Hospital General de L'Hospitalet, Complex Hospitalari Universitari Moisès Broggi, Barcelona, Spain
| | - Israel Molina
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Spain; Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
| | - Clara Crespillo-Andújar
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Spain; Unidad de Patología Importada y Salud Internacional. Hospital Universitario La Paz, Madrid, Spain; National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - Montserrat Gállego
- Barcelona Institute for Global Health, ISGlobal-Hospital Clinic, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Spain; Parasitology Section, Department of Biology, Health, and Environment, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Cristina Ballart
- Barcelona Institute for Global Health, ISGlobal-Hospital Clinic, Universitat de Barcelona, Barcelona, Spain; Parasitology Section, Department of Biology, Health, and Environment, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Juan Carlos Ramirez
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, CONICET-GCBA, Buenos Aires, Argentina
| | - Tilman Aden
- Institute of Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Kenneth Pfarr
- Institute of Medical Microbiology, Immunology, and Parasitology, University Hospital Bonn, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Michel Vaillant
- Competence Centre for Methodology and Statistics, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Tayná Marques
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil
| | - Jayme Fernandes
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil
| | - Bethania Blum
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil
| | - Isabela Ribeiro
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil; Centro de Investigaciones en Epidemiología y Salud Pública (CIESP-CONICET), Buenos Aires, Argentina
| | | | - Joaquim Gascón
- Barcelona Institute for Global Health, ISGlobal-Hospital Clinic, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Spain
| |
Collapse
|
18
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
19
|
Kwakye-Nuako G, Middleton CE, McCall LI. Small molecule mediators of host-T. cruzi-environment interactions in Chagas disease. PLoS Pathog 2024; 20:e1012012. [PMID: 38457443 PMCID: PMC10923493 DOI: 10.1371/journal.ppat.1012012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Abstract
Small molecules (less than 1,500 Da) include major biological signals that mediate host-pathogen-microbiome communication. They also include key intermediates of metabolism and critical cellular building blocks. Pathogens present with unique nutritional needs that restrict pathogen colonization or promote tissue damage. In parallel, parts of host metabolism are responsive to immune signaling and regulated by immune cascades. These interactions can trigger both adaptive and maladaptive metabolic changes in the host, with microbiome-derived signals also contributing to disease progression. In turn, targeting pathogen metabolic needs or maladaptive host metabolic changes is an important strategy to develop new treatments for infectious diseases. Trypanosoma cruzi is a single-celled eukaryotic pathogen and the causative agent of Chagas disease, a neglected tropical disease associated with cardiac and intestinal dysfunction. Here, we discuss the role of small molecules during T. cruzi infection in its vector and in the mammalian host. We integrate these findings to build a theoretical interpretation of how maladaptive metabolic changes drive Chagas disease and extrapolate on how these findings can guide drug development.
Collapse
Affiliation(s)
- Godwin Kwakye-Nuako
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Caitlyn E. Middleton
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, United States of America
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, United States of America
| |
Collapse
|
20
|
de Sousa AS, Vermeij D, Ramos AN, Luquetti AO. Chagas disease. Lancet 2024; 403:203-218. [PMID: 38071985 DOI: 10.1016/s0140-6736(23)01787-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 01/15/2024]
Abstract
Chagas disease persists as a global public health problem due to the high morbidity and mortality burden. Despite the possibility of a cure and advances in transmission control, epidemiological transformations, such as urbanisation and globalisation, and the emerging importance of oral and vertical transmission mean that Chagas disease should be considered an emerging disease, with new cases occurring worldwide. Important barriers to diagnosis, treatment, and care remain, resulting in repressed numbers of reported cases, which in turn leads to inadequate public policies. The validation of new diagnostic tools and treatment options is needed, as existing tools pose serious limitations to access to health care. Integrated models of surveillance, with community and intersectional participation, embedded in the concept of One Health, are essential for control. In addition, mitigation strategies for the main social determinants of health, including difficulties imposed by migration, are important to improve access to comprehensive health care in a globalised scenario.
Collapse
Affiliation(s)
- Andréa Silvestre de Sousa
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Department of Internal Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Debbie Vermeij
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alberto Novaes Ramos
- Department of Community Health, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Alejandro O Luquetti
- Center of Studies for Chagas Disease, Hospital das Clínicas, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
21
|
Desale H, Herrera C, Dumonteil E. Trypanosoma cruzi amastigote transcriptome analysis reveals heterogenous populations with replicating and dormant parasites. Microbes Infect 2024; 26:105240. [PMID: 37866547 DOI: 10.1016/j.micinf.2023.105240] [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: 09/03/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Trypanosoma cruzi is a protozoan parasite causing Chagas disease, with a complex life cycle involving different stages in insect vectors and mammalian hosts. Amastigotes are an intracellular form that replicates in the cytoplasm of host cells, and recent studies suggested that dormant forms may be contributing to parasite persistence, suggesting cellular heterogeneity among amastigotes. We investigated here if a transcriptomic approach could identify some heterogeneity in intracellular amastigotes and identify a dormant population. We used gene expression data derived from bulk RNA-sequencing of T. cruzi infection of human fibroblasts for deconvolution using CDSeq, which allows to simultaneously estimate amastigote cell-type proportions and cell-type-specific expression profiles. Six amastigote subpopulations were identified, confirming intracellular amastigotes heterogeneity, and one population presented characteristics of non-replicative dormant parasites, based on replication markers and TcRAD51 expression. Transcriptomic approaches appear to be powerful to understand T. cruzi cell differentiation and expansion of these studies could provide further insight on the role different cell types in parasite persistence and Chagas disease pathogenesis.
Collapse
Affiliation(s)
- Hans Desale
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
22
|
Zingales B, Macedo AM. Fifteen Years after the Definition of Trypanosoma cruzi DTUs: What Have We Learned? Life (Basel) 2023; 13:2339. [PMID: 38137940 PMCID: PMC10744745 DOI: 10.3390/life13122339] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Trypanosoma cruzi, the protozoan causative of Chagas disease (ChD), exhibits striking genetic and phenotypic intraspecific diversity, along with ecoepidemiological complexity. Human-pathogen interactions lead to distinct clinical presentations of ChD. In 2009, an international consensus classified T. cruzi strains into six discrete typing units (DTUs), TcI to TcVI, later including TcBat, and proposed reproducible genotyping schemes for DTU identification. This article aims to review the impact of classifying T. cruzi strains into DTUs on our understanding of biological, ecoepidemiological, and pathogenic aspects of T. cruzi. We will explore the likely origin of DTUs and the intrinsic characteristics of each group of strains concerning genome organization, genomics, and susceptibility to drugs used in ChD treatment. We will also provide an overview of the association of DTUs with mammalian reservoirs, and summarize the geographic distribution, and the clinical implications, of prevalent specific DTUs in ChD patients. Throughout this review, we will emphasize the crucial roles of both parasite and human genetics in defining ChD pathogenesis and chemotherapy outcome.
Collapse
Affiliation(s)
- Bianca Zingales
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, São Paulo, Brazil
| | - Andréa M. Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil;
| |
Collapse
|
23
|
González S, Wall RJ, Thomas J, Braillard S, Brunori G, Díaz IC, Cantizani J, Carvalho S, Castañeda Casado P, Chatelain E, Cotillo I, Fiandor JM, Francisco AF, Grimsditch D, Keenan M, Kelly JM, Kessler A, Luise C, Lyon JJ, MacLean L, Marco M, Martin JJ, Martinez MS, Paterson C, Read KD, Santos-Villarejo A, Zuccotto F, Wyllie S, Miles TJ, De Rycker M. Short-course combination treatment for experimental chronic Chagas disease. Sci Transl Med 2023; 15:eadg8105. [PMID: 38091410 PMCID: PMC7615676 DOI: 10.1126/scitranslmed.adg8105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects millions of people in the Americas and across the world, leading to considerable morbidity and mortality. Current treatment options, benznidazole (BNZ) and nifurtimox, offer limited efficacy and often lead to adverse side effects because of long treatment durations. Better treatment options are therefore urgently required. Here, we describe a pyrrolopyrimidine series, identified through phenotypic screening, that offers an opportunity to improve on current treatments. In vitro cell-based washout assays demonstrate that compounds in the series are incapable of killing all parasites; however, combining these pyrrolopyrimidines with a subefficacious dose of BNZ can clear all parasites in vitro after 5 days. These findings were replicated in a clinically predictive in vivo model of chronic Chagas disease, where 5 days of treatment with the combination was sufficient to prevent parasite relapse. Comprehensive mechanism of action studies, supported by ligand-structure modeling, show that compounds from this pyrrolopyrimidine series inhibit the Qi active site of T. cruzi cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Knowledge of the molecular target enabled a cascade of assays to be assembled to evaluate selectivity over the human cytochrome b homolog. As a result, a highly selective and efficacious lead compound was identified. The combination of our lead compound with BNZ rapidly clears T. cruzi parasites, both in vitro and in vivo, and shows great potential to overcome key issues associated with currently available treatments.
Collapse
Affiliation(s)
- Silvia González
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Richard J. Wall
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - John Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | | | - Juan Cantizani
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Sandra Carvalho
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | - Ignacio Cotillo
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Jose M. Fiandor
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | | | | | - John M. Kelly
- London School for Hygiene and Tropical Medicine, London, UK
| | - Albane Kessler
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Chiara Luise
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | - Lorna MacLean
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Maria Marco
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - J. Julio Martin
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | - Christy Paterson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Kevin D. Read
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | - Fabio Zuccotto
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Tim J. Miles
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Manu De Rycker
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| |
Collapse
|
24
|
Dean DA, Roach J, Ulrich vonBargen R, Xiong Y, Kane SS, Klechka L, Wheeler K, Jimenez Sandoval M, Lesani M, Hossain E, Katemauswa M, Schaefer M, Harris M, Barron S, Liu Z, Pan C, McCall LI. Persistent Biofluid Small-Molecule Alterations Induced by Trypanosoma cruzi Infection Are Not Restored by Parasite Elimination. ACS Infect Dis 2023; 9:2173-2189. [PMID: 37883691 PMCID: PMC10842590 DOI: 10.1021/acsinfecdis.3c00261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi (T. cruzi) protozoa, is a complicated parasitic illness with inadequate medical measures for diagnosing infection and monitoring treatment success. To address this gap, we analyzed changes in the metabolome of T. cruzi-infected mice via liquid chromatography tandem mass spectrometry of clinically accessible biofluids: saliva, urine, and plasma. Urine was the most indicative of infection status across mouse and parasite genotypes. Metabolites perturbed by infection in urine include kynurenate, acylcarnitines, and threonylcarbamoyladenosine. Based on these results, we sought to implement urine as a tool for the assessment of CD treatment success. Strikingly, it was found that mice with parasite clearance following benznidazole antiparasitic treatment had an overall urine metabolome comparable to that of mice that failed to clear parasites. These results provide a complementary hypothesis to explain clinical trial data in which benznidazole treatment did not improve patient outcomes in late-stage disease, even in patients with successful parasite clearance. Overall, this study provides insights into new small-molecule-based CD diagnostic methods and a new approach to assess functional responses to treatment.
Collapse
Affiliation(s)
- Danya A. Dean
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Jarrod Roach
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | | | - Yi Xiong
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Shelley S. Kane
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - London Klechka
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Kate Wheeler
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | | | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Ekram Hossain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Mitchelle Katemauswa
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Miranda Schaefer
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Morgan Harris
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Sayre Barron
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Chongle Pan
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| |
Collapse
|
25
|
Jayawardhana S, Ward AI, Francisco AF, Lewis MD, Taylor MC, Kelly JM, Olmo F. Benznidazole treatment leads to DNA damage in Trypanosoma cruzi and the persistence of rare widely dispersed non-replicative amastigotes in mice. PLoS Pathog 2023; 19:e1011627. [PMID: 37956215 PMCID: PMC10681306 DOI: 10.1371/journal.ppat.1011627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Benznidazole is the front-line drug used to treat infections with Trypanosoma cruzi, the causative agent of Chagas disease. However, for reasons that are unknown, treatment failures are common. When we examined parasites that survived benznidazole treatment in mice using highly sensitive in vivo and ex vivo bioluminescence imaging, we found that recrudescence is not due to persistence of parasites in a specific organ or tissue that preferentially protects them from drug activity. Surviving parasites are widely distributed and located in host cells where the vast majority contained only one or two amastigotes. Therefore, infection relapse does not arise from a small number of intact large nests. Rather, persisters are either survivors of intracellular populations where co-located parasites have been killed, or amastigotes in single/low-level infected cells exist in a state where they are less susceptible to benznidazole. To better assess the nature of parasite persisters, we exposed infected mammalian cell monolayers to a benznidazole regimen that reduces the intracellular amastigote population to <1% of the pre-treatment level. Of host cells that remained infected, as with the situation in vivo, the vast majority contained only one or two surviving intracellular amastigotes. Analysis, based on non-incorporation of the thymidine analogue EdU, revealed these surviving parasites to be in a transient non-replicative state. Furthermore, treatment with benznidazole led to widespread parasite DNA damage. When the small number of parasites which survive in mice after non-curative treatment were assessed using EdU labelling, this revealed that these persisters were also initially non-replicative. A possible explanation could be that triggering of the T. cruzi DNA damage response pathway by the activity of benznidazole metabolites results in exit from the cell cycle as parasites attempt DNA repair, and that metabolic changes associated with non-proliferation act to reduce drug susceptibility. Alternatively, a small percentage of the parasite population may pre-exist in this non-replicative state prior to treatment.
Collapse
Affiliation(s)
- Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alexander I. Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
26
|
Liu Z, Ulrich vonBargen R, Kendricks AL, Wheeler K, Leão AC, Sankaranarayanan K, Dean DA, Kane SS, Hossain E, Pollet J, Bottazzi ME, Hotez PJ, Jones KM, McCall LI. Localized cardiac small molecule trajectories and persistent chemical sequelae in experimental Chagas disease. Nat Commun 2023; 14:6769. [PMID: 37880260 PMCID: PMC10600178 DOI: 10.1038/s41467-023-42247-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
Post-infectious conditions present major health burdens but remain poorly understood. In Chagas disease (CD), caused by Trypanosoma cruzi parasites, antiparasitic agents that successfully clear T. cruzi do not always improve clinical outcomes. In this study, we reveal differential small molecule trajectories between cardiac regions during chronic T. cruzi infection, matching with characteristic CD apical aneurysm sites. Incomplete, region-specific, cardiac small molecule restoration is observed in animals treated with the antiparasitic benznidazole. In contrast, superior restoration of the cardiac small molecule profile is observed for a combination treatment of reduced-dose benznidazole plus an immunotherapy, even with less parasite burden reduction. Overall, these results reveal molecular mechanisms of CD treatment based on simultaneous effects on the pathogen and on host small molecule responses, and expand our understanding of clinical treatment failure in CD. This link between infection and subsequent persistent small molecule perturbation broadens our understanding of infectious disease sequelae.
Collapse
Affiliation(s)
- Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Rebecca Ulrich vonBargen
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | - Kate Wheeler
- Department of Biology, University of Oklahoma, Norman, OK, USA
| | - Ana Carolina Leão
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Krithivasan Sankaranarayanan
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Danya A Dean
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Shelley S Kane
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Ekram Hossain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Jeroen Pollet
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kathryn M Jones
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA.
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA.
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA.
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA, USA.
| |
Collapse
|
27
|
Fesser A, Beilstein S, Kaiser M, Schmidt RS, Mäser P. Trypanosoma cruzi STIB980: A TcI Strain for Drug Discovery and Reverse Genetics. Pathogens 2023; 12:1217. [PMID: 37887733 PMCID: PMC10610277 DOI: 10.3390/pathogens12101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Since the first published genome sequence of Trypanosoma cruzi in 2005, there have been tremendous technological advances in genomics, reverse genetics, and assay development for this elusive pathogen. However, there is still an unmet need for new and better drugs to treat Chagas disease. Here, we introduce a T. cruzi assay strain that is useful for drug research and basic studies of host-pathogen interactions. T. cruzi STIB980 is a strain of discrete typing unit TcI that grows well in culture as axenic epimastigotes or intracellular amastigotes. We evaluated the optimal parameters for genetic transfection and constructed derivatives of T. cruzi STIB980 that express reporter genes for fluorescence- or bioluminescence-based drug efficacy testing, as well as a Cas9-expressing line for CRISPR/Cas9-mediated gene editing. The genome of T. cruzi STIB980 was sequenced by combining short-read Illumina with long-read Oxford Nanopore technologies. The latter served as the primary assembly and the former to correct mistakes. This resulted in a high-quality nuclear haplotype assembly of 28 Mb in 400 contigs, containing 10,043 open-reading frames with a median length of 1077 bp. We believe that T. cruzi STIB980 is a useful addition to the antichagasic toolbox and propose that it can serve as a DTU TcI reference strain for drug efficacy testing.
Collapse
Affiliation(s)
- Anna Fesser
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Sabina Beilstein
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Remo S Schmidt
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Department Medical Parasitology and Infection Biology, 4123 Allschwil, Switzerland
- University of Basel, 4001 Basel, Switzerland
| |
Collapse
|
28
|
Schijman AG. Unveiling challenges in real-time PCR strategies for detecting treatment failure: observations from clinical trials on chronic Chagas disease. FRONTIERS IN PARASITOLOGY 2023; 2:1260224. [PMID: 39816840 PMCID: PMC11732123 DOI: 10.3389/fpara.2023.1260224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/04/2023] [Indexed: 01/18/2025]
Abstract
Chagas disease (CD) caused by Trypanosoma cruzi remains a Neglected Tropical Disease with limited access to diagnosis and treatment, particularly for chronically infected patients. Clinical trials are underway to improve treatment using new drugs or different regimens, and Real-Time PCR is used to assess the parasitological response as a surrogate biomarker. However, PCR-based strategies have limitations due to the complex nature of T. cruzi infection. The parasite exhibits asynchronous replication, different strains and clones, and diverse tissue tropism, making it challenging to determine optimal timeline points for monitoring treatment response. This mini-review explores factors that affect PCR-based monitoring and summarizes the endpoints used in clinical trials for detecting treatment failure. Serial sampling and cumulative PCR results may improve sensitivity in detecting parasitemia and treatment failure in these trials.
Collapse
Affiliation(s)
- Alejandro G. Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular Dr Héctor N. Torres (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
29
|
Pardo-Rodriguez D, Lasso P, Santamaría-Torres M, Cala MP, Puerta CJ, Méndez Arteaga JJ, Robles J, Cuervo C. Clethra fimbriata hexanic extract triggers alteration in the energy metabolism in epimastigotes of Trypanosoma cruzi. Front Mol Biosci 2023; 10:1206074. [PMID: 37818099 PMCID: PMC10561390 DOI: 10.3389/fmolb.2023.1206074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Chagas disease (ChD), caused by Trypanosoma cruzi, is endemic in American countries and an estimated 8 million people worldwide are chronically infected. Currently, only two drugs are available for therapeutic use against T. cruzi and their use is controversial due to several disadvantages associated with side effects and low compliance with treatment. Therefore, there is a need to search for new tripanocidal agents. Natural products have been considered a potential innovative source of effective and selective agents for drug development to treat T. cruzi infection. Recently, our research group showed that hexanic extract from Clethra fimbriata (CFHEX) exhibits anti-parasitic activity against all stages of T. cruzi parasite, being apoptosis the main cell death mechanism in both epimastigotes and trypomastigotes stages. With the aim of deepening the understanding of the mechanisms of death induced by CFHEX, the metabolic alterations elicited after treatment using a multiplatform metabolomics analysis (RP/HILIC-LC-QTOF-MS and GC-QTOF-MS) were performed. A total of 154 altered compounds were found significant in the treated parasites corresponding to amino acids (Arginine, threonine, cysteine, methionine, glycine, valine, proline, isoleucine, alanine, leucine, glutamic acid, and serine), fatty acids (stearic acid), glycerophospholipids (phosphatidylcholine, phosphatidylethanolamine and phosphatidylserine), sulfur compounds (trypanothione) and carboxylic acids (pyruvate and phosphoenolpyruvate). The most affected metabolic pathways were mainly related to energy metabolism, which was found to be decrease during the evaluated treatment time. Further, exogenous compounds of the triterpene type (betulinic, ursolic and pomolic acid) previously described in C. fimbriata were found inside the treated parasites. Our findings suggest that triterpene-type compounds may contribute to the activity of CFHEX by altering essential processes in the parasite.
Collapse
Affiliation(s)
- Daniel Pardo-Rodriguez
- Grupo de Enfermedades Infecciosas, Pontificia Universidad Javeriana, Bogotá, Colombia
- Grupo de Fitoquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
- Grupo de Productos Naturales, Universidad del Tolima, Tolima, Colombia
- Metabolomics Core Facility—MetCore, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Paola Lasso
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Mary Santamaría-Torres
- Metabolomics Core Facility—MetCore, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Mónica P. Cala
- Metabolomics Core Facility—MetCore, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Concepción J. Puerta
- Grupo de Enfermedades Infecciosas, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Jorge Robles
- Grupo de Fitoquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Claudia Cuervo
- Grupo de Enfermedades Infecciosas, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
30
|
Tarannum A, Rodríguez-Almonacid CC, Salazar-Bravo J, Karamysheva ZN. Molecular Mechanisms of Persistence in Protozoan Parasites. Microorganisms 2023; 11:2248. [PMID: 37764092 PMCID: PMC10534552 DOI: 10.3390/microorganisms11092248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Protozoan parasites are known for their remarkable capacity to persist within the bodies of vertebrate hosts, which frequently results in prolonged infections and the recurrence of diseases. Understanding the molecular mechanisms that underlie the event of persistence is of paramount significance to develop innovative therapeutic approaches, given that these pathways still need to be thoroughly elucidated. The present article provides a comprehensive overview of the latest developments in the investigation of protozoan persistence in vertebrate hosts. The focus is primarily on the function of persisters, their formation within the host, and the specific molecular interactions between host and parasite while they persist. Additionally, we examine the metabolomic, transcriptional, and translational changes that protozoan parasites undergo during persistence within vertebrate hosts, focusing on major parasites such as Plasmodium spp., Trypanosoma spp., Leishmania spp., and Toxoplasma spp. Key findings of our study suggest that protozoan parasites deploy several molecular and physiological strategies to evade the host immune surveillance and sustain their persistence. Furthermore, some parasites undergo stage differentiation, enabling them to acclimate to varying host environments and immune challenges. More often, stressors such as drug exposure were demonstrated to impact the formation of protozoan persisters significantly. Understanding the molecular mechanisms regulating the persistence of protozoan parasites in vertebrate hosts can reinvigorate our current insights into host-parasite interactions and facilitate the development of more efficacious disease therapeutics.
Collapse
Affiliation(s)
| | | | | | - Zemfira N. Karamysheva
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (A.T.); (C.C.R.-A.); (J.S.-B.)
| |
Collapse
|
31
|
Moreira CHV, Bierrenbach AL, Taconeli CA, de Oliveira-da Silva LC, Buss LF, Keating SM, Manuli ER, Carvalho NB, Guastini C, Coco SB, Lindoso JÂL, Franco LAM, Ghilardi F, Sales FCDS, Contestable P, Di Germanio C, Busch MP, Sabino EC. Parasitemia and antibody response to benznidazole treatment in a cohort of patients with chronic Chagas disease. FRONTIERS IN PARASITOLOGY 2023; 2:1235925. [PMID: 39816837 PMCID: PMC11731785 DOI: 10.3389/fpara.2023.1235925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/01/2023] [Indexed: 01/18/2025]
Abstract
Background Evaluating the effectiveness of Chagas disease treatment poses challenges due to the lack of biomarkers for disease progression and therapeutic response. In this study, we aimed to assess the clearance of Trypanosoma cruzi (T. cruzi) parasites in a group of benznidazole (BNZ)-treated chronic Chagas disease patients using high-sensitivity quantitative PCR (qPCR) and track T. cruzi antibody levels through a semiquantitative chemiluminescent assay. Methods A total of 102 T. cruzi seropositive patients with previous PCR-positive results were enrolled in the study. We collected samples 30 days before treatment (T-30d), on the day before initiating BNZ treatment (T0d), and at follow-up visits 60 days (T60d), 6 months (T6M), 12 months (T12M), and 36 months (T36M) after treatment initiation. Treatment efficacy was assessed by testing of serial samples using a target-capture qPCR assay specific to satellite T. cruzi DNA and the ORTHO T. cruzi ELISA Test System for antibody quantitation. Results Of the enrolled individuals, 87 completed at least 50% of the treatment course, and 86 had PCR results at follow-up visits T6M, T12M, and T36M. PCR results exhibited fluctuations before and after treatment, but levels were significantly lower post-treatment. Only 15 cases consistently tested PCR-negative across all post-treatment visits. Notably, nearly all participants demonstrated a declining antibody trajectory, with patients who tested PCR-negative at T36M exhibiting an earlier and more pronounced decline compared to PCR-positive cases at the same visit. Conclusion Our study suggests that serial PCR results pose challenges in interpretation. In contrast, serial antibody levels may serve as an ancillary, or even a more reliable indicator of parasite decline following BNZ treatment. Monitoring antibody levels can provide valuable insights into the efficacy of treatment and the persistence of parasites in Chagas disease patients.
Collapse
Affiliation(s)
- Carlos Henrique Valente Moreira
- Institute of Infectology “Emilio Ribas”, São Paulo, São Paulo, Brazil
- Division of HIV, Infectious Diseases, & Global Medicine- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | | | | | - Léa Campos de Oliveira-da Silva
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Lewis F. Buss
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Centre for Academic Primary Care, University of Bristol, Bristol, United Kingdom
| | | | - Erika Regina Manuli
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Hospital das Clinicas de São Paulo, São Paulo, Brazil
| | | | | | - Sonia Bakkour Coco
- Vitalant Research Institute, San Francisco, CA, United States
- Grifols Diagnostic Solutions, Emeryville, CA, United States
| | - José Ângelo Lauletta Lindoso
- Institute of Infectology “Emilio Ribas”, São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Lucas Augusto Moyses Franco
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Fabio Ghilardi
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Flavia Cristina da Silva Sales
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | | | | | - Michael P. Busch
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Ester Cerdeira Sabino
- Laboratório de Parasitologia Médica (LIM-46), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
32
|
Flores A, Alonso-Vega C, Hermann E, Torrico MC, Montaño Villarroel NA, Torrico F, Carlier Y, Truyens C. Monocytes from Uninfected Neonates Born to Trypanosoma cruzi-Infected Mothers Display Upregulated Capacity to Produce TNF-α and to Control Infection in Association with Maternally Transferred Antibodies. Pathogens 2023; 12:1103. [PMID: 37764911 PMCID: PMC10536721 DOI: 10.3390/pathogens12091103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Activated monocytes/macrophages that produce inflammatory cytokines and nitric oxide are crucial for controlling Trypanosoma cruzi infection. We previously showed that uninfected newborns from T. cruzi infected mothers (M+B- newborns) were sensitized to produce higher levels of inflammatory cytokines than newborns from uninfected mothers (M-B- newborns), suggesting that their monocytes were more activated. Thus, we wondered whether these cells might help limit congenital infection. We investigated this possibility by studying the activation status of M+B- cord blood monocytes and their ability to control T. cruzi in vitro infection. We showed that M+B- monocytes have an upregulated capacity to produce the inflammatory cytokine TNF-α and a better ability to control T. cruzi infection than M-B- monocytes. Our study also showed that T. cruzi-specific Abs transferred from the mother play a dual role by favoring trypomastigote entry into M+B- monocytes and inhibiting intracellular amastigote multiplication. These results support the possibility that some M+B- fetuses may eliminate the parasite transmitted in utero from their mothers, thus being uninfected at birth.
Collapse
Affiliation(s)
- Amilcar Flores
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | - Cristina Alonso-Vega
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | - Emmanuel Hermann
- Laboratory of Parasitology, Faculty of Medicine, ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
| | - Mary-Cruz Torrico
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | | | - Faustino Torrico
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | - Yves Carlier
- Laboratory of Parasitology, Faculty of Medicine, ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
| |
Collapse
|
33
|
Jamabo M, Mahlalela M, Edkins AL, Boshoff A. Tackling Sleeping Sickness: Current and Promising Therapeutics and Treatment Strategies. Int J Mol Sci 2023; 24:12529. [PMID: 37569903 PMCID: PMC10420020 DOI: 10.3390/ijms241512529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the extracellular protozoan parasite Trypanosoma brucei, and targeted for eradication by 2030. The COVID-19 pandemic contributed to the lengthening of the proposed time frame for eliminating human African trypanosomiasis as control programs were interrupted. Armed with extensive antigenic variation and the depletion of the B cell population during an infectious cycle, attempts to develop a vaccine have remained unachievable. With the absence of a vaccine, control of the disease has relied heavily on intensive screening measures and the use of drugs. The chemotherapeutics previously available for disease management were plagued by issues such as toxicity, resistance, and difficulty in administration. The approval of the latest and first oral drug, fexinidazole, is a major chemotherapeutic achievement for the treatment of human African trypanosomiasis in the past few decades. Timely and accurate diagnosis is essential for effective treatment, while poor compliance and resistance remain outstanding challenges. Drug discovery is on-going, and herein we review the recent advances in anti-trypanosomal drug discovery, including novel potential drug targets. The numerous challenges associated with disease eradication will also be addressed.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Maduma Mahlalela
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Adrienne L. Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Centre (BioBRU), Rhodes University, Makhanda 6139, South Africa;
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| |
Collapse
|
34
|
Pereira PML, Fernandes BT, dos Santos VR, Cabral WRC, Lovo-Martins MI, Alonso L, Lancheros CAC, de Paula JC, Camargo PG, Suzukawa HT, Alonso A, Macedo F, Nakamura CV, Tavares ER, de Lima Ferreira Bispo M, Yamauchi LM, Pinge-Filho P, Yamada-Ogatta SF. Antiprotozoal Activity of Benzoylthiourea Derivatives against Trypanosoma cruzi: Insights into Mechanism of Action. Pathogens 2023; 12:1012. [PMID: 37623972 PMCID: PMC10457850 DOI: 10.3390/pathogens12081012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
For decades, only two nitroheterocyclic drugs have been used as therapeutic agents for Chagas disease. However, these drugs present limited effectiveness during the chronic phase, possess unfavorable pharmacokinetic properties, and induce severe adverse effects, resulting in low treatment adherence. A previous study reported that N-(cyclohexylcarbamothioyl) benzamide (BTU-1), N-(tert-butylcarbamothioyl) benzamide (BTU-2), and (4-bromo-N-(3-nitrophenyl) carbamothioyl benzamide (BTU-3) present selective antiprotozoal activity against all developmental forms of Trypanosoma cruzi Y strain. In this study, we investigated the mechanism of action of these compounds through microscopy and biochemical analyses. Transmission electron microscopy analysis showed nuclear disorganization, changes in the plasma membrane with the appearance of blebs and extracellular arrangements, intense vacuolization, mitochondrial swelling, and formation of myelin-like structures. Biochemical results showed changes in the mitochondrial membrane potential, reactive oxygen species content, lipid peroxidation, and plasma membrane fluidity. In addition, the formation of autophagic vacuoles was observed. These findings indicate that BTU-1, BTU-2, and BTU-3 induced profound morphological, ultrastructural, and biochemical alterations in epimastigote forms, triggering an autophagic-dependent cell death pathway.
Collapse
Affiliation(s)
- Patrícia Morais Lopes Pereira
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Bruna Terci Fernandes
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Vitória Ribeiro dos Santos
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Weslei Roberto Correia Cabral
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Maria Isabel Lovo-Martins
- Laboratory of Experimental Immunopathology, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina 86057-970, Brazil;
| | - Lais Alonso
- Institute of Physics, Federal University of Goiás, Goiania 74690-900, Brazil; (L.A.); (A.A.)
| | | | | | - Priscila Goes Camargo
- Laboratory of Medicinal Molecules Synthesis, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil; (P.G.C.); (M.d.L.F.B.)
| | - Helena Tiemi Suzukawa
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Antônio Alonso
- Institute of Physics, Federal University of Goiás, Goiania 74690-900, Brazil; (L.A.); (A.A.)
| | - Fernando Macedo
- Laboratory of Medicinal Molecules Synthesis, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil; (P.G.C.); (M.d.L.F.B.)
| | - Celso Vataru Nakamura
- Laboratory of Technological Innovation in the Development of Drugs and Cosmetics, Department of Basic Health Sciences, State University of Maringá, Maringa 87020-900, Brazil;
| | - Eliandro Reis Tavares
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Marcelle de Lima Ferreira Bispo
- Laboratory of Medicinal Molecules Synthesis, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil; (P.G.C.); (M.d.L.F.B.)
| | - Lucy Megumi Yamauchi
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Phileno Pinge-Filho
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Experimental Immunopathology, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina 86057-970, Brazil;
| | - Sueli Fumie Yamada-Ogatta
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| |
Collapse
|
35
|
Porta EOJ, Kalesh K, Steel PG. Navigating drug repurposing for Chagas disease: advances, challenges, and opportunities. Front Pharmacol 2023; 14:1233253. [PMID: 37576826 PMCID: PMC10416112 DOI: 10.3389/fphar.2023.1233253] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Chagas disease is a vector-borne illness caused by the protozoan parasite Trypanosoma cruzi (T. cruzi). It poses a significant public health burden, particularly in the poorest regions of Latin America. Currently, there is no available vaccine, and chemotherapy has been the traditional treatment for Chagas disease. However, the treatment options are limited to just two outdated medicines, nifurtimox and benznidazole, which have serious side effects and low efficacy, especially during the chronic phase of the disease. Collectively, this has led the World Health Organization to classify it as a neglected disease. To address this problem, new drug regimens are urgently needed. Drug repurposing, which involves the use of existing drugs already approved for the treatment of other diseases, represents an increasingly important option. This approach offers potential cost reduction in new drug discovery processes and can address pharmaceutical bottlenecks in the development of drugs for Chagas disease. In this review, we discuss the state-of-the-art of drug repurposing approaches, including combination therapy with existing drugs, to overcome the formidable challenges associated with treating Chagas disease. Organized by original therapeutic area, we describe significant recent advances, as well as the challenges in this field. In particular, we identify candidates that exhibit potential for heightened efficacy and reduced toxicity profiles with the ultimate objective of accelerating the development of new, safe, and effective treatments for Chagas disease.
Collapse
Affiliation(s)
| | - Karunakaran Kalesh
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Darlington, United Kingdom
| | - Patrick G. Steel
- Department of Chemistry, Durham University, Durham, United Kingdom
| |
Collapse
|
36
|
Robledo SM, Pérez-Silanes S, Fernández-Rubio C, Poveda A, Monzote L, González VM, Alonso-Collado P, Carrión J. Neglected Zoonotic Diseases: Advances in the Development of Cell-Penetrating and Antimicrobial Peptides against Leishmaniosis and Chagas Disease. Pathogens 2023; 12:939. [PMID: 37513786 PMCID: PMC10383258 DOI: 10.3390/pathogens12070939] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
In 2020, the WHO established the road map for neglected tropical diseases 2021-2030, which aims to control and eradicate 20 diseases, including leishmaniosis and Chagas disease. In addition, since 2015, the WHO has been developing a Global Action Plan on Antimicrobial Resistance. In this context, the achievement of innovative strategies as an alternative to replace conventional therapies is a first-order socio-sanitary priority, especially regarding endemic zoonoses in poor regions, such as those caused by Trypanosoma cruzi and Leishmania spp. infections. In this scenario, it is worth highlighting a group of natural peptide molecules (AMPs and CPPs) that are promising strategies for improving therapeutic efficacy against these neglected zoonoses, as they avoid the development of toxicity and resistance of conventional treatments. This review presents the novelties of these peptide molecules and their ability to cross a whole system of cell membranes as well as stimulate host immune defenses or even serve as vectors of molecules. The efforts of the biotechnological sector will make it possible to overcome the limitations of antimicrobial peptides through encapsulation and functionalization methods to obtain approval for these treatments to be used in clinical programs for the eradication of leishmaniosis and Chagas disease.
Collapse
Affiliation(s)
- Sara M. Robledo
- Programa de Estudio y Control de Enfermedades Tropicales PECET, Facultad de Medicina, Universidad de Antioquia, Medellín 050010, Colombia;
| | - Silvia Pérez-Silanes
- Department of Pharmaceutical Technology and Chemistry, ISTUN Instituto de Salud Tropical, IdiSNA, Universidad de Navarra, 31008 Pamplona, Spain;
| | - Celia Fernández-Rubio
- Department of Microbiology and Parasitology, ISTUN Instituto de Salud Tropical, IdiSNA, Universidad de Navarra, 31008 Pamplona, Spain;
| | - Ana Poveda
- DNA Replication and Genome Instability Unit, Grupo de Investigación en Biodiversidad, Zoonosis y Salud Pública (GIBCIZ), Instituto de Investigación en Zoonosis-CIZ, Facultad de Ciencias Químicas, Universidad Central del Ecuador, Quito 170521, Ecuador;
| | - Lianet Monzote
- Department of Parasitology, Institute of Tropical Medicine “Pedro Kourí”, Apartado Postal No. 601, Marianao 13, La Habana 10400, Cuba;
| | - Víctor M. González
- Grupo de Aptámeros, Departamento de Bioquímica-Investigación, IRYCIS-Hospital Universitario Ramón y Cajal, Carretera de Colmenar Viejo Km. 9.100, 28034 Madrid, Spain;
| | - Paloma Alonso-Collado
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Javier Carrión
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
37
|
Gabaldón-Figueira JC, Martinez-Peinado N, Escabia E, Ros-Lucas A, Chatelain E, Scandale I, Gascon J, Pinazo MJ, Alonso-Padilla J. State-of-the-Art in the Drug Discovery Pathway for Chagas Disease: A Framework for Drug Development and Target Validation. Res Rep Trop Med 2023; 14:1-19. [PMID: 37337597 PMCID: PMC10277022 DOI: 10.2147/rrtm.s415273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
Chagas disease is the most important protozoan infection in the Americas, and constitutes a significant public health concern throughout the world. Development of new medications against its etiologic agent, Trypanosoma cruzi, has been traditionally slow and difficult, lagging in comparison with diseases caused by other kinetoplastid parasites. Among the factors that explain this are the incompletely understood mechanisms of pathogenesis of T. cruzi infection and its complex set of interactions with the host in the chronic stage of the disease. These demand the performance of a variety of in vitro and in vivo assays as part of any drug development effort. In this review, we discuss recent breakthroughs in the understanding of the parasite's life cycle and their implications in the search for new chemotherapeutics. For this, we present a framework to guide drug discovery efforts against Chagas disease, considering state-of-the-art preclinical models and recently developed tools for the identification and validation of molecular targets.
Collapse
Affiliation(s)
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Elisa Escabia
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - María-Jesús Pinazo
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| |
Collapse
|
38
|
Bustamante JM, White BE, Wilkerson GK, Hodo CL, Auckland LD, Wang W, McCain S, Hamer SA, Saunders AB, Tarleton RL. Frequency Variation and Dose Modification of Benznidazole Administration for the Treatment of Trypanosoma cruzi Infection in Mice, Dogs, and Nonhuman Primates. Antimicrob Agents Chemother 2023; 67:e0013223. [PMID: 37039666 PMCID: PMC10190575 DOI: 10.1128/aac.00132-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/22/2023] [Indexed: 04/12/2023] Open
Abstract
Trypanosoma cruzi naturally infects a broad range of mammalian species and frequently results in the pathology that has been most extensively characterized in human Chagas disease. Currently employed treatment regimens fail to achieve parasitological cure of T. cruzi infection in the majority of cases. In this study, we have extended our previous investigations of more effective, higher dose, intermittent administration protocols using the FDA-approved drug benznidazole (BNZ), in experimentally infected mice and in naturally infected dogs and nonhuman primates (NHP). Collectively, these studies demonstrate that twice-weekly administration of BNZ for more than 4 months at doses that are ~2.5-fold that of previously used daily dosing protocols, provided the best chance to obtain parasitological cure. Dosing less frequently or for shorter time periods was less dependable in all species. Prior treatment using an ineffective dosing regimen in NHPs did not prevent the attainment of parasitological cure with an intensified BNZ dosing protocol. Furthermore, parasites isolated after a failed BNZ treatment showed nearly identical susceptibility to BNZ as those obtained prior to treatment, confirming the low risk of induction of drug resistance with BNZ and the ability to adjust the treatment protocol when an initial regimen fails. These results provide guidance for the use of BNZ as an effective treatment for T. cruzi infection and encourage its wider use, minimally in high value dogs and at-risk NHP, but also potentially in humans, until better options are available.
Collapse
Affiliation(s)
- Juan M. Bustamante
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Brooke E. White
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Gregory K. Wilkerson
- MD Anderson Cancer Center, Michale E. Keeling Center for Comparative Medicine and Research, Bastrop, Texas, USA
| | - Carolyn L. Hodo
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Research, Texas A&M University, College Station, Texas, USA
| | - Lisa D. Auckland
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Research, Texas A&M University, College Station, Texas, USA
| | - Wei Wang
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | | | - Sarah A. Hamer
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Research, Texas A&M University, College Station, Texas, USA
| | - Ashley B. Saunders
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Rick L. Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
39
|
Domagalska MA, Barrett MP, Dujardin JC. Drug resistance in Leishmania: does it really matter? Trends Parasitol 2023; 39:251-259. [PMID: 36803859 DOI: 10.1016/j.pt.2023.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/19/2023]
Abstract
Treatment failure (TF) jeopardizes the management of parasitic diseases, including leishmaniasis. From the parasite's point of view, drug resistance (DR) is generally considered as central to TF. However, the link between TF and DR, as measured by in vitro drug susceptibility assays, is unclear, some studies revealing an association between treatment outcome and drug susceptibility, others not. Here we address three fundamental questions aiming to shed light on these ambiguities. First, are the right assays being used to measure DR? Second, are the parasites studied, which are generally those that adapt to in vitro culture, actually appropriate? Finally, are other parasite factors - such as the development of quiescent forms that are recalcitrant to drugs - responsible for TF without DR?
Collapse
Affiliation(s)
| | - Michael P Barrett
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
40
|
Rao SPS, Manjunatha UH, Mikolajczak S, Ashigbie PG, Diagana TT. Drug discovery for parasitic diseases: powered by technology, enabled by pharmacology, informed by clinical science. Trends Parasitol 2023; 39:260-271. [PMID: 36803572 DOI: 10.1016/j.pt.2023.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 02/22/2023]
Abstract
While prevention is a bedrock of public health, innovative therapeutics are needed to complement the armamentarium of interventions required to achieve disease control and elimination targets for neglected diseases. Extraordinary advances in drug discovery technologies have occurred over the past decades, along with accumulation of scientific knowledge and experience in pharmacological and clinical sciences that are transforming many aspects of drug R&D across disciplines. We reflect on how these advances have propelled drug discovery for parasitic infections, focusing on malaria, kinetoplastid diseases, and cryptosporidiosis. We also discuss challenges and research priorities to accelerate discovery and development of urgently needed novel antiparasitic drugs.
Collapse
Affiliation(s)
| | | | | | - Paul G Ashigbie
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA.
| | | |
Collapse
|
41
|
Tarleton RL. Effective drug discovery in Chagas disease. Trends Parasitol 2023; 39:423-431. [PMID: 37024318 DOI: 10.1016/j.pt.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
The Chagas field has gone >50 years without tangible progress toward new therapies. My colleagues and I have recently reported on a benzoxaborole compound that achieves consistent parasitological cure in experimentally infected mice and in naturally infected non-human primates (NHPs). While these results do not assure success in human clinical trials, they significantly de-risk this process and form a strong justification for such trials. Highly effective drug discovery depends on a solid understanding of host and parasite biology and excellent knowledge in designing and validating chemical entities. This opinion piece seeks to provide perspectives on the process that led to the discovery of AN15368, with the hope that this will facilitate the discovery of additional clinical candidates for Chagas disease.
Collapse
Affiliation(s)
- Rick L Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
42
|
Rojas-Pirela M, Kemmerling U, Quiñones W, Michels PAM, Rojas V. Antimicrobial Peptides (AMPs): Potential Therapeutic Strategy against Trypanosomiases? Biomolecules 2023; 13:biom13040599. [PMID: 37189347 DOI: 10.3390/biom13040599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Trypanosomiases are a group of tropical diseases that have devastating health and socio-economic effects worldwide. In humans, these diseases are caused by the pathogenic kinetoplastids Trypanosoma brucei, causing African trypanosomiasis or sleeping sickness, and Trypanosoma cruzi, causing American trypanosomiasis or Chagas disease. Currently, these diseases lack effective treatment. This is attributed to the high toxicity and limited trypanocidal activity of registered drugs, as well as resistance development and difficulties in their administration. All this has prompted the search for new compounds that can serve as the basis for the development of treatment of these diseases. Antimicrobial peptides (AMPs) are small peptides synthesized by both prokaryotes and (unicellular and multicellular) eukaryotes, where they fulfill functions related to competition strategy with other organisms and immune defense. These AMPs can bind and induce perturbation in cell membranes, leading to permeation of molecules, alteration of morphology, disruption of cellular homeostasis, and activation of cell death. These peptides have activity against various pathogenic microorganisms, including parasitic protists. Therefore, they are being considered for new therapeutic strategies to treat some parasitic diseases. In this review, we analyze AMPs as therapeutic alternatives for the treatment of trypanosomiases, emphasizing their possible application as possible candidates for the development of future natural anti-trypanosome drugs.
Collapse
|
43
|
Impact of Laboratory-Adapted Intracellular Trypanosoma cruzi Strains on the Activity Profiles of Compounds with Anti- T. cruzi Activity. Microorganisms 2023; 11:microorganisms11020476. [PMID: 36838441 PMCID: PMC9967867 DOI: 10.3390/microorganisms11020476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Chagas disease is caused by infection with the protozoan parasite, Trypanosoma cruzi. The disease causes ~12,000 deaths annually and is one of the world's 20 neglected tropical diseases, as defined by the World Health Organisation. The drug discovery pipeline for Chagas disease currently has few new clinical candidates, with high attrition rates an ongoing issue. To determine if the Trypanosoma cruzi strain utilised to assess in vitro compound activity impacts activity, a comparison of laboratory-adapted T. cruzi strains from differing geographical locations was undertaken for a selection of compounds with anti-T. cruzi activity. To minimise the possible effect of differences in experimental methodology, the same host cell and multiplicity of infection were utilised. To determine whether the compound exposure time influenced results, activity was determined following exposure for 48 and 72 h of incubation. To ascertain whether replication rates affected outcomes, comparative rates of replication of the T. cruzi strains were investigated, using the nucleoside analogue, 5-ethynyl-2'-deoxyuridine. Minimal differences in the in vitro activity of compounds between strains were observed following 48 h incubation, whereas significant differences were observed following 72 h incubation, in particular for the cytochrome P450 inhibitors tested and the cell cycle inhibitor, camptothecin. Thus, the use of panels of laboratory adapted strains in vitro may be dependent on the speed of action that is prioritised. For the identification of fast-acting compounds, an initial shorter duration assay using a single strain may be used. A longer incubation to identify compound activity may alternatively require profiling of compounds against multiple T. cruzi strains.
Collapse
|
44
|
Bustamante JM, White BE, Wilkerson GK, Hodo CL, Auckland LD, Wang W, McCain S, Hamer SA, Saunders AB, Tarleton RL. Frequency variation and dose modification of benznidazole administration for the treatment of Trypanosoma cruzi infection in mice, dogs and non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526739. [PMID: 36778432 PMCID: PMC9915573 DOI: 10.1101/2023.02.01.526739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Trypanosoma cruzi naturally infects a broad range of mammalian species and frequently results in the pathology that has been most extensively characterized in human Chagas disease. Currently employed treatment regimens fail to achieve parasitological cure of T. cruzi infection in the majority of cases. In this study, we have extended our previous investigations of more effective, higher dose, intermittent administration protocols using the FDA-approved drug benznidazole (BNZ), in experimentally infected mice and in naturally infected dogs and non-human primates (NHP). Collectively these studies demonstrate that twice-weekly administration of BNZ for more than 4 months at doses that are ∼2.5-fold that of previously used daily dosing protocols, provided the best chance to obtain parasitological cure. Dosing less frequently or for shorter time periods was less dependable in all species. Prior treatment using an ineffective dosing regimen in NHPs did not prevent the attainment of parasitological cure with an intensified BNZ dosing protocol. Furthermore, parasites isolated after a failed BNZ treatment showed nearly identical susceptibility to BNZ as those obtained prior to treatment, confirming the low risk of induction of drug resistance with BNZ and the ability to adjust the treatment protocol when an initial regimen fails. These results provide guidance for the use of BNZ as an effective treatment for T. cruzi infection and encourage its wider use, minimally in high value dogs and at-risk NHP, but also potentially in humans, until better options are available.
Collapse
Affiliation(s)
- Juan M Bustamante
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Brooke E White
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Gregory K Wilkerson
- MD Anderson Cancer Center, Michale E. Keeling Center for Comparative Medicine and Research, Bastrop, Texas, USA
| | - Carolyn L Hodo
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Research, Texas A&M University, College Station, Texas, USA
| | - Lisa D Auckland
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Research, Texas A&M University, College Station, Texas, USA
| | - Wei Wang
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | | | - Sarah A Hamer
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Research, Texas A&M University, College Station, Texas, USA
| | - Ashley B Saunders
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Rick L Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
45
|
Development and Application of an Assay to Evaluate the Anti-Parasitic Effect of Humoral Responses against Trypanosoma cruzi. Microorganisms 2023; 11:microorganisms11020241. [PMID: 36838206 PMCID: PMC9966445 DOI: 10.3390/microorganisms11020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Mounting a balanced and robust humoral immune response is of utmost importance for reducing the infectivity of Trypanosoma cruzi. While the role of such a response in controlling the infection is well known, there is a lack of tools that can be used to quickly evaluate it. We developed a serum parasite inhibition assay (to evaluate changes in the parasite infection after exposing infective T. cruzi trypomastigotes to serum samples from infected patients). It is based on Vero cells as the hosts and the Tulahuen β-galactosidase parasite strain, genetically engineered to be quantifiable by spectrophotometry. In parallel, we developed an in-house ELISA to correlate the anti-T. cruzi antibody titres of the clinical samples with their observed anti-parasitic effect in the serum parasite inhibition assay. Serum samples from chronically T. cruzi-infected patients significantly inhibited parasite invasion in a titre-dependant manner, regardless of the patient's clinical status, compared to samples from the non-infected controls. In addition, there was a clear correlation between the reactivity of the samples to the whole-parasite lysates by ELISA and the inhibitory effect. The results of this work confirm the previously described anti-parasitic effect of the serum of individuals exposed to T. cruzi and present a framework for its large-scale evaluation in further studies. The serum parasite inhibition assay represents a reproducible way to evaluate the intensity and anti-parasitic effect of humoral responses against T. cruzi, which could be applied to the evaluation of candidate antigens/epitopes in the design of Chagas disease vaccine candidates.
Collapse
|
46
|
Abstract
Leishmaniasis (visceral and cutaneous), Chagas disease and human African trypanosomiasis cause substantial death and morbidity, particularly in low- and middle-income countries. Although the situation has improved for human African trypanosomiasis, there remains an urgent need for new medicines to treat leishmaniasis and Chagas disease; the clinical development pipeline is particularly sparse for Chagas disease. In this Review, we describe recent advances in our understanding of the biology of the causative pathogens, particularly from the drug discovery perspective, and we explore the progress that has been made in the development of new drug candidates and the identification of promising molecular targets. We also explore the challenges in developing new clinical candidates and discuss potential solutions to overcome such hurdles.
Collapse
|
47
|
Identification of Aryl Polyamines Derivatives as Anti- Trypanosoma cruzi Agents Targeting Iron Superoxide Dismutase. Pharmaceutics 2022; 15:pharmaceutics15010140. [PMID: 36678771 PMCID: PMC9863987 DOI: 10.3390/pharmaceutics15010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
Chagas disease (CD) is a tropical and potentially fatal infection caused by Trypanosoma cruzi. Although CD was limited to Latin America as a silent disease, CD has become widespread as a result of globalization. Currently, 6-8 million people are infected worldwide, and no effective treatment is available. Here, we identify new effective agents against T. cruzi. In short, 16 aryl polyamines were screened in vitro against different T. cruzi strains, and lead compounds were evaluated in vivo after oral administration in both the acute and chronic infections. The mode of action was also evaluated at the energetic level, and its high activity profile could be ascribed to a mitochondria-dependent bioenergetic collapse and redox stress by inhibition of the Fe-SOD enzyme. We present compound 15 as a potential compound that provides a step forward for the development of new agents to combat CD.
Collapse
|
48
|
Kamiya T, Paton DG, Catteruccia F, Reece SE. Targeting malaria parasites inside mosquitoes: ecoevolutionary consequences. Trends Parasitol 2022; 38:1031-1040. [PMID: 36209032 PMCID: PMC9815470 DOI: 10.1016/j.pt.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/05/2022]
Abstract
Proof-of-concept studies demonstrate that antimalarial drugs designed for human treatment can also be applied to mosquitoes to interrupt malaria transmission. Deploying a new control tool is ideally undertaken within a stewardship programme that maximises a drug's lifespan by minimising the risk of resistance evolution and slowing its spread once emerged. We ask: what are the epidemiological and evolutionary consequences of targeting parasites within mosquitoes? Our synthesis argues that targeting parasites inside mosquitoes (i) can be modelled by readily expanding existing epidemiological frameworks; (ii) provides a functionally novel control method that has potential to be more robust to resistance evolution than targeting parasites in humans; and (iii) could extend the lifespan and clinical benefit of antimalarials used exclusively to treat humans.
Collapse
Affiliation(s)
- Tsukushi Kamiya
- Centre for Interdisciplinary Research in Biology, Collège de France, Paris, France; HRB Clinical Research Facility, National University of Ireland, Galway, Ireland; Institute of Ecology and Evolution, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
| | - Douglas G Paton
- Department of Immunology and Infectious Disease, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Disease, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA
| | - Sarah E Reece
- Institute of Ecology and Evolution, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
49
|
Fiuza LFDA, Batista DGJ, Girão RD, Hulpia F, Finamore-Araújo P, Aldfer MM, Elmahallawy EK, De Koning HP, Moreira O, Van Calenbergh S, Soeiro MDNC. Phenotypic Evaluation of Nucleoside Analogues against Trypanosoma cruzi Infection: In Vitro and In Vivo Approaches. Molecules 2022; 27:molecules27228087. [PMID: 36432189 PMCID: PMC9695592 DOI: 10.3390/molecules27228087] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi (T. cruzi), is a serious public health problem. Current treatment is restricted to two drugs, benznidazole and nifurtimox, displaying serious efficacy and safety drawbacks. Nucleoside analogues represent a promising alternative as protozoans do not biosynthesize purines and rely on purine salvage from the hosts. Protozoan transporters often present different substrate specificities from mammalian transporters, justifying the exploration of nucleoside analogues as therapeutic agents. Previous reports identified nucleosides with potent trypanocidal activity; therefore, two 7-derivatized tubercidins (FH11706, FH10714) and a 3′-deoxytubercidin (FH8513) were assayed against T. cruzi. They were highly potent and selective, and the uptake of the tubercidin analogues appeared to be mediated by the nucleoside transporter TcrNT2. At 10 μM, the analogues reduced parasitemia >90% in 2D and 3D cardiac cultures. The washout assays showed that FH10714 sterilized the infected cultures. Given orally, the compounds did not induce noticeable mouse toxicity (50 mg/kg), suppressed the parasitemia of T. cruzi-infected Swiss mice (25 mg/kg, 5 days) and presented DNA amplification below the limit of detection. These findings justify further studies with longer treatment regimens, as well as evaluations in combination with nitro drugs, aiming to identify more effective and safer therapies for Chagas disease.
Collapse
Affiliation(s)
- Ludmila F. de A. Fiuza
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, 4365 Manguinhos, Rio de Janeiro 21040-360, Brazil
| | - Denise G. J. Batista
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, 4365 Manguinhos, Rio de Janeiro 21040-360, Brazil
| | - Roberson D. Girão
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, 4365 Manguinhos, Rio de Janeiro 21040-360, Brazil
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Paula Finamore-Araújo
- Laboratório de Virologia Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro 20000-000, Brazil
| | - Mustafa M. Aldfer
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow 62694, UK
| | - Ehab Kotb Elmahallawy
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow 62694, UK
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - Harry P. De Koning
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow 62694, UK
| | - Otacílio Moreira
- Laboratório de Virologia Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro 20000-000, Brazil
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Maria de Nazaré C. Soeiro
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, 4365 Manguinhos, Rio de Janeiro 21040-360, Brazil
- Correspondence: ; Tel.: +55-21-2562-1368
| |
Collapse
|
50
|
Pandey RP, Nascimento MS, Franco CH, Bortoluci K, Silva MN, Zingales B, Gibaldi D, Castaño Barrios L, Lannes-Vieira J, Cariste LM, Vasconcelos JR, Moraes CB, Freitas-Junior LH, Kalil J, Alcântara L, Cunha-Neto E. Drug Repurposing in Chagas Disease: Chloroquine Potentiates Benznidazole Activity against Trypanosoma cruzi In Vitro and In Vivo. Antimicrob Agents Chemother 2022; 66:e0028422. [PMID: 36314800 PMCID: PMC9664849 DOI: 10.1128/aac.00284-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Drug combinations and drug repurposing have emerged as promising strategies to develop novel treatments for infectious diseases, including Chagas disease. In this study, we aimed to investigate whether the repurposed drugs chloroquine (CQ) and colchicine (COL), known to inhibit Trypanosoma cruzi infection in host cells, could boost the anti-T. cruzi effect of the trypanocidal drug benznidazole (BZN), increasing its therapeutic efficacy while reducing the dose needed to eradicate the parasite. The combination of BZN and COL exhibited cytotoxicity to infected cells and low antiparasitic activity. Conversely, a combination of BZN and CQ significantly reduced T. cruzi infection in vitro, with no apparent cytotoxicity. This effect seemed to be consistent across different cell lines and against both the partially BZN-resistant Y and the highly BZN-resistant Colombiana strains. In vivo experiments in an acute murine model showed that the BZN+CQ combination was eight times more effective in reducing T. cruzi infection in the acute phase than BZN monotherapy. In summary, our results demonstrate that the concomitant administration of CQ and BZN potentiates the trypanocidal activity of BZN, leading to a reduction in the dose needed to achieve an effective response. In a translational context, it could represent a higher efficacy of treatment while also mitigating the adverse effects of high doses of BZN. Our study also reinforces the relevance of drug combination and repurposing approaches in the field of Chagas disease drug discovery.
Collapse
Affiliation(s)
- Ramendra P. Pandey
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Marilda Savoia Nascimento
- School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute of Tropical Medicine of São Paulo, São Paulo, Brazil
| | - Caio Haddad Franco
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karina Bortoluci
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
| | - Marcelo Nunes Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Bianca Zingales
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Leonardo Moro Cariste
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
| | - Jose Ronnie Vasconcelos
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
- Department of Microbiology, Immunology, and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Carolina Borsoi Moraes
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lucio H. Freitas-Junior
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Laura Alcântara
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute of Tropical Medicine of São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|