Next Article in Journal
Role of Sinorhizobium meliloti and Escherichia coli Long-Chain Acyl-CoA Synthetase FadD in Long-Term Survival
Previous Article in Journal
Pseudomonas PS01 Isolated from Maize Rhizosphere Alters Root System Architecture and Promotes Plant Growth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

High-Throughput Screening of the ReFRAME Library Identifies Potential Drug Repurposing Candidates for Trypanosoma cruzi

by
Jean A. Bernatchez
1,2,
Emily Chen
3,
Mitchell V. Hull
3,
Case W. McNamara
3,
James H. McKerrow
1,2 and
Jair L. Siqueira-Neto
1,2,*
1
Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA 92093, USA
2
Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, CA 92093, USA
3
Calibr, a Division of The Scripps Research Institute, La Jolla, CA 92037, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(4), 472; https://doi.org/10.3390/microorganisms8040472
Submission received: 19 February 2020 / Revised: 21 March 2020 / Accepted: 23 March 2020 / Published: 26 March 2020
(This article belongs to the Section Parasitology)

Abstract

:
Chagas disease, caused by the kinetoplastid parasite Trypanosoma cruzi, affects between 6 and 7 million people worldwide, with an estimated 300,000 to 1 million of these cases in the United States. In the chronic phase of infection, T. cruzi can cause severe gastrointestinal and cardiac disease, which can be fatal. Currently, only benznidazole is clinically approved by the FDA for pediatric use to treat this infection in the USA. Toxicity associated with this compound has driven the search for new anti-Chagas agents. Drug repurposing is a particularly attractive strategy for neglected diseases, as pharmacological parameters and toxicity are already known for these compounds, reducing costs and saving time in the drug development pipeline. Here, we screened 7680 compounds from the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library, a collection of drugs or compounds with confirmed clinical safety, against T. cruzi. We identified seven compounds of interest with potent in vitro activity against the parasite with a therapeutic index of 10 or greater, including the previously unreported activity of the antiherpetic compound 348U87. These results provide the framework for further development of new T. cruzi leads that can potentially move quickly to the clinic.

1. Introduction

Trypanosoma cruzi, the causative agent of Chagas disease, is a protozoan parasite that is primarily transmitted to humans via triatomine insects (known as kissing bugs) during a blood meal. Infection by T. cruzi manifests initially in an acute phase of infection, and if left untreated, proceeds to a chronic phase [1]. During the acute phase, mild or unremarkable symptoms such as fever, fatigue, rash, headache, or swelling at the site of the triatome bite may present. When left untreated, the primary infection usually resolves in weeks, but residual parasites remain in the host’s body, evolving to the chronic phase. Over the span of years to decades, approximately 30% of those infected individuals will manifest cardiac and/or gastrointestinal complications, leading to morbidity and mortality [2].
Current treatment options are very limited for Chagas disease; only benznidazole is clinically approved for pediatric use in the case of acute T. cruzi infections in the United States. Benznidazole and nifurtimox are available off-label via the CDC for compassionate use for all other cases of this infection. However, severe side effects associated with the use of these medications lead to high levels of patient discontinuation of treatment. Furthermore, the usefulness of benznidazole in the chronic phase of infection is disputed within the Chagas research community [3,4,5].
Limited effort from the pharmaceutical industry to develop a medication for T. cruzi infections further complicates progress towards anti-Chagas agents better than benznidazole and nifurtimox. Rising costs and high levels of failure of drug molecules in clinical trials, due to adverse events and lack of efficacy, present further general barriers to the development of medications. One cost-effective strategy involves repurposing existing drugs with known toxicity and pharmacokinetic profiles for other indications [6]. This has the potential to speed up drug development efforts, reduce costs, and lower the chance of adverse events presenting in clinical trials. The Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library, a comprehensive set of molecules with tested clinical safety, has been previously used to identify potential drug repurposing hits for neglected tropical diseases [7,8]. In this work, we screened 7680 compounds from this library against the medically relevant intracellular, amastigote form of T. cruzi, infecting mouse myoblasts using a high-throughput, phenotypic cellular imaging assay that our group has successfully used in previous studies to identify novel antitrypanosomal agents [9,10,11]. We identified seven compounds with suitable selectivity indexes (SIs) for drug repurposing; two of these, the antiherpetic drug 348U87 and the serotonin receptor binder 3-[4-[4-(2-Methoxyphenyl)piperazine-1-yl]butyl]-6-[2-[4-(4-fluorobenzoyl)piperidine-1-yl]ethyl]benzothiazole-2(3H)-one, have not previously been reported as anti-Chagas compounds, and may target the parasite through a novel mechanism. These molecules form an attractive collection of lead molecules for potential drug repurposing to treat Chagas disease.

2. Materials and Methods

2.1. Cells

C2C12 mouse myoblasts (ATCC CRL-1772) and CA-I/72 T. cruzi (kindly donated by J. Dvorak, NIH) were cultured in Dulbecco’s Modified Eagle Medium (Invitrogen, Carlsbad, CA, USA), supplemented with 5% fetal bovine serum (Sigma Aldrich, St. Louis, MO, USA) and 1% penicillin-streptomycin (Invitrogen, Carlsbad, CA, USA) at 37 °C and 5% CO2 essentially as described [11]. Passaging of CA-I/72 T. cruzi was conducted weekly via co-culture with C2C12 host cells.

2.2. Phenotypic Imaging Assay

Compounds from the ReFRAME library, benznidazole (Sigma Alderich, St. Louis, MO, USA) and DMSO (Sigma Alderich, St. Louis, MO, USA), were transferred to black 1536-well plates (Greiner Bio One, Kremsmünster, Austria) with clear bottoms using an Acoustic Transfer System (ATS) instrument (EDC Biosystems, Fremont, CA, USA). C2C12 cells were seeded at a density of 100 cells per well, and CA-I/72 T. cruzi parasites were seeded at a density of 1500 cells per well, using a Multidrop Combi liquid handler (Thermo Scientific, Waltham, MA, USA). Plates were incubated at 37 °C and 5% CO2 for 72 h in humidified trays to reduce edge effect. Following this incubation, paraformaldehyde (4% final concentration) in 1× phosphate buffered saline (PBS, Invitrogen, 10010023) was used to fix the cells for 1 h. The cells were then subsequently treated with 5 µg/mL DAPI staining solution (Sigma Aldrich, D9542) for 1 h. Next, the plates were imaged using an ImageXpress Micro XLS automated high-content imager (Molecular Devices, San Jose, CA, USA), using the 10× fluorescence objective. Images were analyzed automatically using a custom image analysis module [9,11].

2.3. Software

Chemical structures were prepared using ChemDraw Professional 18.1 (Perkin Elmer, Waltham, MA, USA). EC50 and CC50 values were generated using GraphPad Prism 8 (GraphPad Software, San Diego, CA, USA).

3. Results

3.1. Primary Screening of the ReFRAME Library against T. cruzi Using a High-Content Imaging Assay

Compounds from the ReFRAME library were pre-spotted on 1536 clear-bottom black well plates in 100% dimethylsulfoxide (DMSO) for a final concentration of 10 µM in 10 µL final volume (and 0.1% DMSO final concentration). C2C12 mouse myoblasts and CA-I/72 strain T. cruzi trypomastigotes were added to the plate in a 1:15 infection ratio and incubated for 72 h at 37 °C and 5% CO2. Cells were then fixed with 4% paraformaldehyde (final concentration), and stained with 5 µg/mL of 4’,6-diamidino-2-phenylindole (DAPI) to highlight the nuclei from the host cells and parasites. Using an ImageXpress Micro XLS automated microscope (10× magnification setting), fluorescence images of C2C12 and T. cruzi amastigote nuclei were acquired for each well, and the number of host cell nuclei and amastigote nuclei were automatically determined using a custom image analysis module as previously described [9,10,11] (Figure 1). Parasite amastigote nuclei were selectively counted over trypomastigote nuclei, via distance gating of parasitic nuclei distance from host cell nuclei in the microscopy images.
Infection levels were calculated as a ratio of the number of T. cruzi amastigotes per C2C12 host cell, as determined by nuclei counting. Compound toxicity was determined by dividing the number of host cell nuclei in a drug-treated well by the average of the vehicle controls. For both infection ratios and cell viability ratios, values were normalized to the vehicle controls to determine percent activity and toxicity, respectively. Control wells containing uninfected C2C12 cells, infected C2C12 cells with 0.1% DMSO, and infected C2C12 cells with 0.1% DMSO and 50 µM benznidazole were prepared in each plate for data normalization. The mean Z’ for the 10 plates tested was 0.52 with a standard error of 0.01. Hit selection cutoffs for the primary screen were set at 70% antiparasitic activity (70% parasite reduction compared to untreated controls) and 50% host cell viability at 10 µM compound compared to untreated controls (represents approximately three standard deviations from the average of the untreated controls). We identified 238 compounds (2% of the total library) that met these selection criteria. A summary of the primary screening data is shown in Figure 2.

3.2. Counter-Screen of 238 Hits in Dose Response

To validate the hits obtained from our primary screen, compounds were re-spotted in duplicate in a 10-point, 3-fold dilution dose response, with 10 µM as the highest concentration of inhibitor. Using the high-content imaging assay, 238 primary hits were retested in duplicate. We identified seven compounds of interest using the following cutoff criteria: at least 70% antiparasitic activity at a given drug concentration in one of the two dose response replicates, and a therapeutic index of 10 or greater. Half maximal effective concentration (EC50), half maximal cytotoxic concentration (CC50), and selective index (SI; defined by the ratio of CC50 to EC50) for validated hits are shown in Table 1, and the chemical structures of these compounds are displayed in Figure 3. The seven compounds retained were NSC-706744, 348U87, ASP-8273, XR 5944, Prenyl-IN-1, 3-[4-[4-(2-Methoxyphenyl)piperazine-1-yl]butyl]-6-[2-[4-(4-fluorobenzoyl)piperidine-1-yl]ethyl]benzothiazole-2(3H)-one and incadronate disodium.

4. Discussion

Given the toxicity of currently available drugs to treat T. cruzi infection, research efforts have been made to explore additional candidate molecules against the parasite. In this work, we screened a drug repurposing library for compounds with anti-Chagasic activity. We identified seven molecules as having potent in vitro activity against T. cruzi and a SI of at least 10 against C2C12 cardiomyocyte host cells.
As validation for our assay, we identified a number of compounds from classes which have previously been reported as having anti-Chagas activity, namely farnesyltransferase inhibitors [12,13,14,15,16] (Prenyl-IN-1, incadronate disodium) and DNA topoisomerase inhibitors [17,18,19,20] (NSC-706744, XR 5944).
We also identified the EGFR inhibitor ASP-8273 (naquotinib) [21,22,23] as an inhibitor of T. cruzi with good potency (EC50 of 2.7 nM) and a SI of 191. Several studies have successfully explored kinase inhibitors of trypanosomatids as therapeutic agents [24,25,26], and this compound may represent yet another possible candidate for repurposing or further chemical derivatization.
Interestingly, we identified two compounds with intriguing primary indications that may target T. cruzi by novel mechanisms. First, serotonin receptor ligands such as 3-[4-[4-(2-Methoxyphenyl)piperazine-1-yl]butyl]-6-[2-[4-(4-fluorobenzoyl)piperidine-1-yl]ethyl]benzothiazole-2(3H)-one [27] have been investigated as agents to treat anxiety and panic disorders. This compound had an EC50 of 22 nM and a SI of 145, making it an attractive candidate for drug repurposing and animal model testing. Second, the herpes virus drug 348U87 (EC50 of 0.63 nM and a SI of 1294), which targets the viral ribonucleotide reductase [28,29,30,31], has been shown to potentiate the activity of acyclovir in topical applications. The thiosemicarbazone iron chelator 3-AP has also been shown to inactivate the ribonucleotide reductase of T. brucei [32], and 348U87 may act against a homologous protein of T. cruzi in a similar manner. For both the aforementioned compounds, mechanistic studies are planned and ongoing to identify the precise molecular targets of these inhibitors.
Follow-up studies in our group will test our most promising compounds in mouse models of T. cruzi infection, to establish proper dosing protocols and in vivo efficacy. In sum, these compounds represent new potent leads with known pharmacological parameters and possibly novel mechanisms of action against T. cruzi, making them attractive candidates for accelerated development as anti-Chagas agents.

Author Contributions

J.A.B., E.C., M.V.H., C.W.M., J.H.M., and J.L.S.-N. conceived and designed the experiments; J.A.B., E.C., and M.V.H. performed experiments; J.A.B., E.C., M.V.H., C.W.M., J.H.M., and J.L.S.-N. analyzed the data; J.A.B. and J.L.S.-N. wrote the original draft of the manuscript; J.A.B., E.C., M.V.H., C.W.M., J.H.M., and J.L.S.-N. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Bill & Melinda Gates Foundation (OPP1107194).

Acknowledgments

The screening experiments were performed at the UCSD Screening Core.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lidani, K.C.F.; Andrade, F.A.; Bavia, L.; Damasceno, F.S.; Beltrame, M.H.; Messias-Reason, I.J.; Sandri, T.L. Chagas disease: From discovery to a worldwide health problem. Front. Public Health 2019, 7, 166. [Google Scholar] [CrossRef] [PubMed]
  2. Prata, A. Clinical and epidemiological aspects of Chagas disease. Lancet Infect. Dis. 2001, 1, 92–100. [Google Scholar] [CrossRef]
  3. Caldas, I.S.; Santos, E.G.; Novaes, R.D. An evaluation of benznidazole as a Chagas disease therapeutic. Expert Opin. Pharmacother. 2019, 20, 1797–1807. [Google Scholar] [CrossRef] [PubMed]
  4. Morillo, C.A.; Marin-Neto, J.A.; Avezum, A.; Sosa-Estani, S.; Rassi, A.; Rosas, F.; Villena, E.; Quiroz, R.; Bonilla, R.; Britto, C.; et al. BENEFIT Investigators Randomized trial of benznidazole for chronic chagas’ cardiomyopathy. N. Engl. J. Med. 2015, 373, 1295–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rassi, A.; Marin, J.A.; Rassi, A. Chronic Chagas cardiomyopathy: A review of the main pathogenic mechanisms and the efficacy of aetiological treatment following the BENznidazole Evaluation for Interrupting Trypanosomiasis (BENEFIT) trial. Memórias Inst. Oswaldo Cruz 2017, 112, 224–235. [Google Scholar] [CrossRef]
  6. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef]
  7. Janes, J.; Young, M.E.; Chen, E.; Rogers, N.H.; Burgstaller-Muehlbacher, S.; Hughes, L.D.; Love, M.S.; Hull, M.V.; Kuhen, K.L.; Woods, A.K.; et al. The ReFRAME library as a comprehensive drug repurposing library and its application to the treatment of cryptosporidiosis. Proc. Natl. Acad. Sci. USA 2018, 115, 10750–10755. [Google Scholar] [CrossRef] [Green Version]
  8. Kim, Y.-J.; Cubitt, B.; Chen, E.; Hull, M.V.; Chatterjee, A.K.; Cai, Y.; Kuhn, J.H.; de la Torre, J.C. The ReFRAME library as a comprehensive drug repurposing library to identify mammarenavirus inhibitors. Antivir. Res. 2019, 169, 104558. [Google Scholar] [CrossRef]
  9. Moon, S.; Siqueira-Neto, J.L.; Moraes, C.B.; Yang, G.; Kang, M.; Freitas-Junior, L.H.; Hansen, M.A.E. An image-based algorithm for precise and accurate high throughput assessment of drug activity against the human parasite Trypanosoma cruzi. PLoS ONE 2014, 9, e87188. [Google Scholar] [CrossRef] [Green Version]
  10. Boudreau, P.D.; Miller, B.W.; McCall, L.-I.; Almaliti, J.; Reher, R.; Hirata, K.; Le, T.; Siqueira-Neto, J.L.; Hook, V.; Gerwick, W.H. Design of Gallinamide A Analogs as Potent Inhibitors of the Cysteine Proteases Human Cathepsin L and Trypanosoma cruzi Cruzain. J. Med. Chem. 2019, 62, 9026–9044. [Google Scholar] [CrossRef]
  11. Ekins, S.; de Siqueira-Neto, J.L.; McCall, L.-I.; Sarker, M.; Yadav, M.; Ponder, E.L.; Kallel, E.A.; Kellar, D.; Chen, S.; Arkin, M.; et al. Machine Learning Models and Pathway Genome Data Base for Trypanosoma cruzi Drug Discovery. PLoS Negl. Trop. Dis. 2015, 9, e0003878. [Google Scholar] [CrossRef] [PubMed]
  12. Kraus, J.M.; Verlinde, C.L.M.J.; Karimi, M.; Lepesheva, G.I.; Gelb, M.H.; Buckner, F.S. Rational modification of a candidate cancer drug for use against Chagas disease. J. Med. Chem. 2009, 52, 1639–1647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kraus, J.M.; Tatipaka, H.B.; McGuffin, S.A.; Chennamaneni, N.K.; Karimi, M.; Arif, J.; Verlinde, C.L.M.J.; Buckner, F.S.; Gelb, M.H. Second generation analogues of the cancer drug clinical candidate tipifarnib for anti-Chagas disease drug discovery. J. Med. Chem. 2010, 53, 3887–3898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rodrígues-Poveda, C.A.; González-Pacanowska, D.; Szajnman, S.H.; Rodríguez, J.B. 2-alkylaminoethyl-1,1-bisphosphonic acids are potent inhibitors of the enzymatic activity of Trypanosoma cruzi squalene synthase. Antimicrob. Agents Chemother. 2012, 56, 4483–4486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Shang, N.; Li, Q.; Ko, T.-P.; Chan, H.-C.; Li, J.; Zheng, Y.; Huang, C.-H.; Ren, F.; Chen, C.-C.; Zhu, Z.; et al. Squalene synthase as a target for Chagas disease therapeutics. PLoS Pathog. 2014, 10, e1004114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Bosc, D.; Mouray, E.; Cojean, S.; Franco, C.H.; Loiseau, P.M.; Freitas-Junior, L.H.; Moraes, C.B.; Grellier, P.; Dubois, J. Highly improved antiparasitic activity after introduction of an N-benzylimidazole moiety on protein farnesyltransferase inhibitors. Eur. J. Med. Chem. 2016, 109, 173–186. [Google Scholar] [CrossRef]
  17. Douc-Rasy, S.; Kayser, A.; Riou, G. A specific inhibitor of type I DNA-topoisomerase of Trypanosoma cruzi: Dimethyl-hydroxy-ellipticinium. Biochem. Biophys. Res. Commun. 1983, 117, 1–5. [Google Scholar] [CrossRef]
  18. Kerschmann, R.L.; Wolfson, J.S.; McHugh, G.L.; Dickersin, G.R.; Hooper, D.C.; Swartz, M.N. Novobiocin-induced ultrastructural changes and antagonism of DNA synthesis in Trypanosoma cruzi amastigotes growing in cell-free medium. J. Protozool. 1989, 36, 14–20. [Google Scholar] [CrossRef]
  19. Gonzales-Perdomo, M.; de Castro, S.L.; Meirelles, M.N.; Goldenberg, S. Trypanosoma cruzi proliferation and differentiation are blocked by topoisomerase II inhibitors. Antimicrob. Agents Chemother. 1990, 34, 1707–1714. [Google Scholar] [CrossRef] [Green Version]
  20. Zuma, A.A.; Cavalcanti, D.P.; Maia, M.C.P.; de Souza, W.; Motta, M.C.M. Effect of topoisomerase inhibitors and DNA-binding drugs on the cell proliferation and ultrastructure of Trypanosoma cruzi. Int. J. Antimicrob. Agents 2011, 37, 449–456. [Google Scholar] [CrossRef] [Green Version]
  21. Yu, H.A.; Spira, A.; Horn, L.; Weiss, J.; West, H.; Giaccone, G.; Evans, T.; Kelly, R.J.; Desai, B.; Krivoshik, A.; et al. A Phase I, Dose Escalation Study of Oral ASP8273 in Patients with Non-small Cell Lung Cancers with Epidermal Growth Factor Receptor Mutations. Clin. Cancer Res. 2017, 23, 7467–7473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hirano, T.; Yasuda, H.; Hamamoto, J.; Nukaga, S.; Masuzawa, K.; Kawada, I.; Naoki, K.; Niimi, T.; Mimasu, S.; Sakagami, H.; et al. Pharmacological and Structural Characterizations of Naquotinib, a Novel Third-Generation EGFR Tyrosine Kinase Inhibitor, in EGFR-Mutated Non-Small Cell Lung Cancer. Mol. Cancer Ther. 2018, 17, 740–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tanaka, H.; Sakagami, H.; Kaneko, N.; Konagai, S.; Yamamoto, H.; Matsuya, T.; Yuri, M.; Yamanaka, Y.; Mori, M.; Takeuchi, M.; et al. Mutant-Selective Irreversible EGFR Inhibitor, Naquotinib, Inhibits Tumor Growth in NSCLC Models with EGFR-Activating Mutations, T790M Mutation, and AXL Overexpression. Mol. Cancer Ther. 2019, 18, 1366–1373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Klug, D.M.; Tschiegg, L.; Díaz González, R.; Rojas-Barros, D.I.; Perez-Moreno, G.; Ceballos-Perez, G.; García-Hernández, R.; Martínez-Martínez, M.S.; Manzano, P.; Ruiz-Perez, L.M.; et al. Hit-to-lead optimization of benzoxazepinoindazoles as human African trypanosomiasis therapeutics. J. Med. Chem. 2019, 63, 2527–2546. [Google Scholar] [CrossRef]
  25. Simões-Silva, M.R.; De Araújo, J.S.; Peres, R.B.; Da Silva, P.B.; Batista, M.M.; De Azevedo, L.D.; Bastos, M.M.; Bahia, M.T.; Boechat, N.; Soeiro, M.N.C. Repurposing strategies for Chagas disease therapy: The effect of imatinib and derivatives against Trypanosoma cruzi. Parasitology 2019, 146, 1006–1012. [Google Scholar] [CrossRef]
  26. Wyllie, S.; Thomas, M.; Patterson, S.; Crouch, S.; De Rycker, M.; Lowe, R.; Gresham, S.; Urbaniak, M.D.; Otto, T.D.; Stojanovski, L.; et al. Cyclin-dependent kinase 12 is a drug target for visceral leishmaniasis. Nature 2018, 560, 192–197. [Google Scholar] [CrossRef]
  27. Diouf, O.; Carato, P.; Lesieur, I.; Rettori, M.; Caignard, D. Synthesis and pharmacological evaluation of novel 4-(4-fluorobenzoyl)piperidine derivatives as mixed 5-HT1A/5-HT2A/D2 receptor ligands. Eur. J. Med. Chem. 1999, 34, 69–73. [Google Scholar] [CrossRef]
  28. Spector, T.; Harrington, J.A.; Porter, D.J. Herpes and human ribonucleotide reductases. Inhibition by 2-acetylpyridine 5-[(2-chloroanilino)-thiocarbonyl]-thiocarbonohydrazone (348U87). Biochem. Pharmacol. 1991, 42, 91–96. [Google Scholar] [CrossRef]
  29. Spector, T.; Lobe, D.C.; Ellis, M.N.; Blumenkopf, T.A.; Szczech, G.M. Inactivators of herpes simplex virus ribonucleotide reductase: Hematological profiles and in vivo potentiation of the antiviral activity of acyclovir. Antimicrob. Agents Chemother. 1992, 36, 934–937. [Google Scholar] [CrossRef] [Green Version]
  30. Safrin, S.; Schacker, T.; Delehanty, J.; Hill, E.; Corey, L. Potential for combined therapy with 348U87, a ribonucleotide reductase inhibitor, and acyclovir as treatment for acyclovir-resistant herpes simplex virus infection. J. Med. Virol. 1993, 1, 146–149. [Google Scholar] [CrossRef]
  31. Safrin, S.; Schacker, T.; Delehanty, J.; Hill, E.; Corey, L. Topical treatment of infection with acyclovir-resistant mucocutaneous herpes simplex virus with the ribonucleotide reductase inhibitor 348U87 in combination with acyclovir. Antimicrob. Agents Chemother. 1993, 37, 975–979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ellis, S.; Sexton, D.W.; Steverding, D. Trypanotoxic activity of thiosemicarbazone iron chelators. Exp. Parasitol. 2015, 150, 7–12. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Automated segmentation analysis of T. cruzi amastigote and C2C12 mouse cardiomyocyte nuclei. (A) Fluorescence microscopy image (10× magnification) of 4’,6-diamidino-2-phenylindole (DAPI)-stained C2C12 infected with CA-I/72 T. cruzi amastigotes 72 h post-infection. (B) Custom module segmentation of host and parasite cell nuclei using MetaXpress 5.0 (Molecular Devices). Host cell nuclei are in yellow and parasite nuclei are in dark blue.
Figure 1. Automated segmentation analysis of T. cruzi amastigote and C2C12 mouse cardiomyocyte nuclei. (A) Fluorescence microscopy image (10× magnification) of 4’,6-diamidino-2-phenylindole (DAPI)-stained C2C12 infected with CA-I/72 T. cruzi amastigotes 72 h post-infection. (B) Custom module segmentation of host and parasite cell nuclei using MetaXpress 5.0 (Molecular Devices). Host cell nuclei are in yellow and parasite nuclei are in dark blue.
Microorganisms 08 00472 g001
Figure 2. Primary screening data for the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library against T. cruzi in the phenotypic high-content imaging assay. Scatter plot of normalized % activity against CA-I/72 T. cruzi (normalized antiparasitic activity %, Y-axis) and the host cell C2C12 viability % (normalized host viability %, X-axis) for the ReFRAME library. The red dots represent the uninfected controls, green dots represent 50 µM benznidazole control (reference drug), blue dots represent untreated controls (0.1% DMSO), and the yellow dots are the tested compounds. A vertical line at 50% normalized host viability and a horizontal line at 70% normalized antiparasitic activity highlight the top right quadrant where the compounds were selected as hits for dose-response confirmation.
Figure 2. Primary screening data for the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library against T. cruzi in the phenotypic high-content imaging assay. Scatter plot of normalized % activity against CA-I/72 T. cruzi (normalized antiparasitic activity %, Y-axis) and the host cell C2C12 viability % (normalized host viability %, X-axis) for the ReFRAME library. The red dots represent the uninfected controls, green dots represent 50 µM benznidazole control (reference drug), blue dots represent untreated controls (0.1% DMSO), and the yellow dots are the tested compounds. A vertical line at 50% normalized host viability and a horizontal line at 70% normalized antiparasitic activity highlight the top right quadrant where the compounds were selected as hits for dose-response confirmation.
Microorganisms 08 00472 g002
Figure 3. Chemical structures of hit compounds from the high-throughput screening campaign.
Figure 3. Chemical structures of hit compounds from the high-throughput screening campaign.
Microorganisms 08 00472 g003
Table 1. EC50 and CC50 values for 7 validated hits from the ReFRAME library against CA-I/72 T. cruzi in the phenotypic high-content imaging assay, ranked in order of potency. Values were calculated from duplicate dose response data, +/− standard error (SE). EC50 values are for T. cruzi CA-I/72 parasites. CC50 values are for C2C12 cardiomyocyte host cells. Selectivity index = CC50/EC50.
Table 1. EC50 and CC50 values for 7 validated hits from the ReFRAME library against CA-I/72 T. cruzi in the phenotypic high-content imaging assay, ranked in order of potency. Values were calculated from duplicate dose response data, +/− standard error (SE). EC50 values are for T. cruzi CA-I/72 parasites. CC50 values are for C2C12 cardiomyocyte host cells. Selectivity index = CC50/EC50.
Compound NameEC50 CA-I/72 T. cruzi (nM)CC50 C2C12 (nM)Selectivity Index (SI)
NSC-7067440.44 +/− 0.0894 +/− 56214
348U870.63 +/− 0.45815 +/− 2151294
ASP-82732.7 +/− 1.9515 +/− 280191
XR 59443.5 +/− 6.846 +/− 2113
Prenyl-IN-118 +/− 12182 +/− 9010
3-[4-[4-(2-Methoxyphenyl)piperazine-1-yl]butyl]-6-[2-[4-(4-fluorobenzoyl)piperidine-1-yl]ethyl]benzothiazole-2(3H)-one22 +/− 233190 +/− 1202145
Incadronate Disodium480 +/− 385>10,000>20

Share and Cite

MDPI and ACS Style

Bernatchez, J.A.; Chen, E.; Hull, M.V.; McNamara, C.W.; McKerrow, J.H.; Siqueira-Neto, J.L. High-Throughput Screening of the ReFRAME Library Identifies Potential Drug Repurposing Candidates for Trypanosoma cruzi. Microorganisms 2020, 8, 472. https://doi.org/10.3390/microorganisms8040472

AMA Style

Bernatchez JA, Chen E, Hull MV, McNamara CW, McKerrow JH, Siqueira-Neto JL. High-Throughput Screening of the ReFRAME Library Identifies Potential Drug Repurposing Candidates for Trypanosoma cruzi. Microorganisms. 2020; 8(4):472. https://doi.org/10.3390/microorganisms8040472

Chicago/Turabian Style

Bernatchez, Jean A., Emily Chen, Mitchell V. Hull, Case W. McNamara, James H. McKerrow, and Jair L. Siqueira-Neto. 2020. "High-Throughput Screening of the ReFRAME Library Identifies Potential Drug Repurposing Candidates for Trypanosoma cruzi" Microorganisms 8, no. 4: 472. https://doi.org/10.3390/microorganisms8040472

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop