Abstract
Parasitic worm infections impose a significant public health burden, affecting over 2 billion people, particularly in low-income regions. The limited efficacy of current treatments highlights the urgent need for new anthelmintic agents. This study investigates the potential antiparasitic activity of 1,10-phenanthroline-5,6-dione (phendione) and its metal complexes, [Cu(phendione)3](ClO4)2.8H2O and [Ag(phendione)2](ClO4), against Schistosoma mansoni, the causative agent of intestinal schistosomiasis, and Angiostrongylus cantonensis, responsible for eosinophilic meningitis in humans. Additionally, the compounds were tested on Caenorhabditis elegans, a model organism for drug discovery. All compounds exhibited strong antiparasitic activity, with Cu-phendione showing the greatest potency (EC50 = 2.3 µM for S. mansoni and 6.4 µM for A. cantonensis). Ag-phendione also demonstrated significant activity, achieving EC₅₀ values of 6.5 µM against S. mansoni and 12.7 µM against A. cantonensis. The lethal dose (LD50) values in C. elegans were over 40 times higher, indicating selective antiparasitic effects. Cytotoxicity assays using Vero cells revealed a low toxicity profile and a high selectivity index. Given the promising biological properties of phendione and its metal complexes, these findings contribute to the growing body of research seeking to address the urgent need for new anthelmintic therapies.
Similar content being viewed by others
Introduction
Parasitic worm infections, or helminthiasis, represent a substantial but often overlooked global health burden. Affecting over 2 billion individuals, primarily in tropical and subtropical regions, these infections disproportionately impact impoverished and marginalized populations with limited access to clean water and sanitation1. Helminthiasis contributes significantly to morbidity and mortality, and many emerging infectious diseases of parasitic origin are zoonotic, further complicating control efforts. Recognizing the urgent need for intervention, the World Health Organization (WHO) has included helminthic diseases in its 2021 roadmap, aiming to eliminate neglected diseases by 20302. Despite ongoing efforts, the arsenal of effective anthelmintic agents remains limited3.
Schistosomiasis, caused by the intravascular trematode of the genus Schistosoma, stands out as one of the most significant parasitic diseases globally, particularly in terms of public health and economic impact. Affecting millions in Africa, the Middle East, South America, and the Caribbean, S. mansoni infection leads to chronic morbidity due to the formation of granulomas and fibrosis from trapped eggs in human tissues4. While praziquantel is the only drug currently used in mass treatment programs5, its limitations, such as reduced efficacy against juvenile parasites and the emergence of drug resistance6, have highlighted the need for alternative therapies7.
Similarly, Angiostrongylus cantonensis, the rat lungworm, presents a growing zoonotic threat8, causing eosinophilic meningitis and other neurological complications in humans9. This nematode, prevalent in mollusk hosts, has spread across various continents, including Africa, Southeast Asia, and the Americas, with cases often linked to travel and misdiagnosed due to the mild symptoms10. Despite its public health importance, there is no effective anthelmintic treatment for A. cantonensis infections, underscoring the need for new therapeutic options11,12.
Transition metal complexes, especially those involving bioactive ligands, have emerged as a promising class of compounds in the search for novel antiparasitic drugs13. These complexes offer impressive chemical diversity and versatility, which depend on the metal of choice, its oxidation state, the number and type of coordinating ligands, and specific magnetic and/or optical properties14. This versatility allows for the rational design of compounds tailored to interact with specific biological targets, addressing the limitations of traditional anthelmintics.
Phenanthrenes, secondary metabolites produced by plants, serve as the chemical backbone for several biologically active compounds15,16. One such example is 1,10-phenanthroline-5,6-dione (phendione), a phenanthrene-based compound, whose metal complexes have shown significant potential due to their unique coordination chemistry and interactions with biological macromolecules17,18. Phendione and its metal complexes have been investigated for their antimicrobial and anticancer properties19,20, establishing them as promising candidates for antiparasitic research21,22. However, their potential against helminths remains largely unexplored.
Given the limited efficacy of current anthelmintic treatments and the pressing need for novel agents, this study investigates the potential antiparasitic properties of phendione and its copper, [Cu(phendione)3](ClO4)2·8H2O – Cu-phendione, and silver, [Ag(phendione)2](ClO4) – Ag-phendione, complexes. We evaluate their in vitro effects on S. mansoni and A. cantonensis and further explore their activity using Caenorhabditis elegans, a model organism for drug discovery. To assess their potential as therapeutic agents, we also performed cytotoxicity assays using Vero cells to determine their selectivity and safety profiles.
Results
The anthelmintic activities of phendione and its metal complexes (Cu-phendione and Ag-phendione) (Fig. 1) were evaluated alongside standard control drugs: praziquantel for S. mansoni, albendazole for A. cantonensis, doxorubicin for cytotoxicity in Vero cells, and ivermectin for C. elegans. Additionally, the simple metal salts AgNO₃ and CuSO₄·5 H₂O were included for comparison (Tables 1and Fig. 2).
Viability of adult Schistosoma mansoni worms after incubation with different concentrations of phendione and its metal complexes (Cu-phendione and Ag-phendione). Control groups included parasites incubated in drug-free medium, praziquantel (PZQ), metal salts (AgNO₃ and CuSO₄·5 H₂O), and 1% DMSO. Schistosomes were obtained from animals by perfusion 49 days post-infection. Each concentration was tested in triplicate, and worm viability was monitored for up to 72 h. Data represent the mean ± S.D. from at least three independent experiments performed in triplicate.
Antiparasitic activity against Schistosoma mansoni
Cu-phendione exhibited the highest potency, with an EC50 of 2.3 µM against S. mansoni, outperforming both phendione (EC50 = 18.8 µM) and Ag-phendione (EC50 = 6.5 µM). While praziquantel remained the most effective control drug (EC50 = 1.2 µM), the potency of Cu-phendione was comparable. In contrast, the simple metal salts AgNO₃ and CuSO₄·5 H₂O showed no antischistosomal activity (Table 1). The negative control group (1% DMSO) displayed no significant alterations, with worms remaining motile throughout the entire incubation period (Fig. 2).
Antiparasitic activity against Angiostrongylus cantonensis L1 larvae
Cu-phendione exhibited strong activity against A. cantonensis (EC50 = 6.4 µM), outperforming phendione (EC50 = 25.6 µM) and demonstrating similar efficacy to albendazole (EC50 = 10.7 µM). Ag-phendione also showed good efficacy (EC50 = 12.7 µM) but was less active than Cu-phendione (Table 1). In contrast, the simple metal salts AgNO₃ and CuSO₄·5 H₂O showed no activity against A. cantonensis. Phendione treatment significantly reduced larval motility compared to the control group, while DMSO-treated larvae maintained normal activity throughout the assay (Fig. 3).
Viability of Angiostrongylus cantonensis L1 larvae during incubation with different concentrations of phendione and its metal complexes (Cu-phendione and Ag-phendione). Control groups included larvae incubated in drug-free medium, albendazole (ALB), metal salts (AgNO₃ and CuSO₄·5 H₂O), and 1% DMSO. Each concentration was tested in triplicate, and larval viability was monitored for up to 24 h. Data represent the mean ± S.D. from at least three independent experiments performed in triplicate.
Toxicity and selectivity index
None of the tested compounds exhibited cytotoxicity in Vero cells at concentrations up to 200 µM (Fig. 4). In comparison, doxorubicin had a CC50 value of 9.6 µM, indicating that phendione and its metal complexes have significantly lower toxicity. The selectivity index (SI) values for Cu-phendione were particularly noteworthy, with SI > 86.9 for S. mansoni and SI > 31.2 for A. cantonensis, indicating high therapeutic potential. Ag-phendione also showed favorable SI values, with SI > 307 for S. mansoni and SI > 15.5 for A. cantonensis (Table 1).
Toxicity of phendione and its metal complexes (Cu-phendione and Ag-phendione) on Vero cells and Caenorhabditis elegans. Doxorubicin and ivermectin were used as reference drugs, while 1% DMSO-treated cells or C. elegans served as controls. Each concentration was tested in triplicate, with viability monitored over 72 h. Data represent the mean ± S.D. from at least three independent experiments performed in triplicate.
The compounds were further evaluated for toxicity in C. elegans (Fig. 4). Cu-phendione exhibited an LD50 of 280 µM, significantly lower than ivermectin (LD50 = 4.1 µM), indicating lower toxicity. Similarly, Ag-phendione showed an LD50 of 167 µM, while phendione demonstrated the least toxicity (LD50 = 719 µM) (Table 1). These results suggest selective toxicity of the metal complexes toward parasitic helminths while minimizing the risk to non-target organisms.
Discussion
This study highlights the potent antiparasitic activity of phendione and its metal complexes, particularly Cu-phendione and Ag-phendione, against S. mansoni and A. cantonensis. These results emphasize the therapeutic potential of metallodrugs in addressing parasitic infections, especially considering the limitations of existing anthelmintic treatments. Importantly, the tested compounds exhibited selective toxicity, showing minimal impact on non-parasitic organisms like C. elegans and low cytotoxicity in Vero cells.
Among the tested compounds, Cu-phendione demonstrated the highest potency, with EC50 values comparable to those of praziquantel for S. mansoni and albendazole for A. cantonensis. Its enhanced efficacy compared to Ag-phendione aligns with previous studies indicating that copper complexes exhibit superior antiparasitic activity, such as against Trichomonas vaginalis21 and Leishmania braziliensis23. Notably, the inactivity of simple metal salts (AgNO₃ and CuSO₄·5 H₂O) reinforces the importance of metal-ligand coordination in optimizing biological activity.
The enhanced performance of Cu-phendione may be attributed to its involvement in parasite metabolism and oxidative stress, as previously reported in studies with S. mansoni24. Recent research has also identified peptidases as potential targets of Cu-phendione in Leishmania23 and T. vaginalis25, while metabolic disruption and altered membrane potential have been observed in Leishmania species treated with Cu-phendione22. These findings suggest that the coordination between the metal center and phendione enhances interactions with key biological targets, increasing the parasites’ susceptibility to treatment. Additionally, oxidative stress response signaling pathways may contribute to nematode death26,27. Further investigations are needed to elucidate the precise mechanisms underlying these interactions and optimize the design of future metallodrugs.
The schistosomicidal activity observed in this study surpasses many plant-derived products reported in the literature. For example, EC50 values reported for S. mansoni adult worms include 12.5 µM for verrucosin28, 26.1 µM for diterpene ent-kaur-16-en-19-oic acid29, 30 µM for neolignan licarin A30, 31.9 µM for dehydrodieugenol B31, 42.16 µM for carvacryl acetate32, 50 µM for cnicin33, licochalcone A34, 2-oxopopulifolic acid methyl ester, and 2-oxopopulifolic acid35, 56.8 µM for monoterpene carvacrol36, and 81.8 µM for flavonoid kaempferol37. Compared to the aforementioned compounds, which are obtained directly from plants, phendiones are obtained by synthesis and are easily obtained in large quantities. The superior performance of phendione-based complexes in this study underscores their potential as more effective alternatives to many of these plant-derived compounds. Additionally, unlike the aforementioned plant-derived compounds, which require extraction and purification processes, phendiones are synthetically produced and can be easily obtained in large quantities, making them more scalable and cost-effective for therapeutic applications.
In addition to their potent antiparasitic activity, the metal complexes exhibited high selectivity. The EC50 values were more than 40 times higher in C. elegans than in parasitic worms, demonstrating selective targeting. Furthermore, the compounds showed low cytotoxicity in Vero cells, with selectivity indices exceeding 86.9 for S. mansoni and 31.2 for A. cantonensis, reinforcing their favorable safety profiles. These findings align with previous studies reporting minimal toxicity of phendione derivatives in various cell lines21,38 and low toxicity in Galleria mellonella larvae38,39, as well as in mammalian models, including mice38 and hamsters40.
Despite the absence of toxicity observed in in vitro and in vivo models with phendione and its metal complexes, it is important to acknowledge that metal-based drugs also present certain limitations that must be addressed to realize their full therapeutic potential. These include concerns about potential toxicity, as metals can accumulate in tissues and cause adverse effects41. Furthermore, the stability of metal-based complexes under physiological conditions and their interactions with off-target biomolecules may compromise their efficacy and safety41,42. Future research should prioritize addressing these limitations through careful optimization of metal-ligand combinations, the development of targeted delivery systems, and comprehensive safety evaluations to ensure their viability as alternative therapeutics for parasitic diseases.
Finally, future research should focus on in vivo studies to validate the efficacy, pharmacokinetics, and safety of these compounds. Investigating potential synergistic effects with established drugs, such as praziquantel or albendazole, could further enhance therapeutic outcomes. Additionally, understanding the mechanisms of action in greater detail may provide insights for developing new metallodrugs with improved selectivity and efficacy. Such advancements are particularly critical in addressing the growing challenge of drug resistance in parasitic infections. Metal-based drugs, with their unique properties and diverse mechanisms, offer a promising alternative to overcome resistance issues, as they can target different biological pathways compared to traditional therapies. This versatility underscores their potential as a valuable addition to the arsenal against drug-resistant parasitic diseases.
Conclusions
This study demonstrates the potent antiparasitic activity of phendione and its metal complexes, particularly Cu-phendione, against two major helminth species. The selective toxicity observed, combined with low cytotoxicity in mammalian cells, underscores the therapeutic promise of these compounds. These findings provide a strong foundation for further in vivo studies and preclinical development, offering new opportunities to expand the limited arsenal of anthelmintic agents. Moreover, exploring synergistic combinations with existing drugs could enhance efficacy and mitigate drug resistance, contributing to more effective treatment strategies for parasitic infections.
Methods
Compounds
1,10-Phenanthroline-5,6-dione (phendione) and its metal complexes, [Ag(phendione)₂]ClO₄ (Ag-phendione) and Cu(phendione)₃(ClO₄)₂·8 H₂O (Cu-phendione) (Fig. 1), were synthesized following the protocols previously described43. Details of the synthesis and characterization of the compounds are provided in the Supplementary Information. These compounds were dissolved in dimethyl sulfoxide (DMSO) and stored at 4 °C until use. Praziquantel, albendazole, and ivermectin were generously provided by Ecovet Indústria Veterinária Ltda (São Paulo, Brazil), while doxorubicin was purchased from Sigma-Aldrich (St. Louis, MO, USA).
Maintenance of parasitic worms
The life cycle of Schistosoma mansoni (Belo Horizonte strain) was maintained at the Research Center on Neglected Diseases, Guarulhos University, using Biomphalaria glabrata snails as the intermediate host and Swiss mice (Mus musculus) as the definitive host. Similarly, the Angiostrongylus cantonensis (NPDN-AC strain) life cycle was maintained by alternating between B. glabrata snails and Wistar rats (Rattus norvegicus). All animals were housed under controlled conditions (22 °C, ~ 50% humidity) with ad libitum access to food and water.
In vitro assay with adult S. mansoni
Adult S. mansoni worms were obtained by hepatic portal vein perfusion from Swiss mice, 49 days post-infection, following established protocols44,45. The worms were washed in RPMI 1640 medium with antibiotics and transferred to 24-well plates, with one pair of adult worms per well. Each well contained 1 mL of RPMI 1640 medium enriched with 10% fetal bovine serum, penicillin (100 U/mL), and streptomycin (100 µg/mL).
Test compounds, praziquantel, and metal salts (AgNO₃ and CuSO₄·5 H₂O) were added at concentrations starting from 50 µM. DMSO (1%) served as the negative control. Plates were incubated at 37 °C with 5% CO₂ for up to 72 h, and worm viability was assessed at 0, 24, 48, and 72 h using an inverted microscope. Parasite death was determined by the absence of movement for at least one minute upon gentle mechanical stimulation46.
In vitro assay with A. cantonensis L1 larvae
First-stage larvae (L1) of A. cantonensis were isolated from the feces of Wistar rats, using the Rugai technique, and washed in RPMI 1640 medium with antibiotics. Approximately 100 larvae per well were transferred to 96-well plates12. The compounds, praziquantel, and metal salts were tested at concentrations starting from 50 µM, with incubation at 21 °C. Larval viability was assessed at 0 and 24 h under an inverted microscope. Movement was categorized as immobile, intermittent, slow, or highly active. Efficacy was defined as at least 60% of larvae becoming immobile within 24 h11,12.
Lethal toxicity assay in C. elegans
The Bristol N2 strain of Caenorhabditis elegans was cultured on nematode growth medium (NGM) plates seeded with Escherichia coli OP5047. L4-stage larvae were synchronized and transferred to 96-well plates containing M9 medium, with 60 larvae per well48. The compounds and ivermectin were tested at concentrations starting from 1,000 µM, with 1% DMSO as the negative control. Viability was assessed after 24 h of incubation at 21 °C by observing movement30. Lethal toxicity was defined as 60% or more of the larvae exhibiting complete immobility49.
Cytotoxicity assay in vero cells
Vero cells (ATCC CCL-81) were maintained in DMEM medium supplemented with 10% fetal bovine serum, penicillin (100 U/mL), and streptomycin (100 µg/mL) at 37 °C with 5% CO₂. The cells were seeded into 96-well plates at 2 × 10³ cells per well and incubated with the test compounds or doxorubicin at concentrations starting from 200 µM, with 1% DMSO as the negative control. Notably, the 200 µM concentration represents the maximal solubility of the compounds under the assay conditions and adheres to standard practices for selectivity index calculations50. Additionally, 1% DMSO was confirmed to be non-toxic to the cells. Cytotoxicity was assessed using the MTT assay51,52. After 72 h, MTT solution was added, followed by 3 h of incubation. Absorbance was measured at 595 nm using an Epoch Microplate Spectrophotometer (BioTek Instruments, Winooski, VT, USA). Assays were performed in triplicate and repeated three times. Selectivity index (SI) values were calculated as the ratio of the 50% cytotoxic concentration (CC50) in Vero cells to the 50% effective concentration (EC50) in the parasitic helminths53.
Data analysis
Statistical analyses were conducted using GraphPad Prism 8.0. EC50, CC50, and LD50 values were determined through sigmoidal dose-response curves48,54. Differences between groups were analyzed using one-way ANOVA followed by Tukey’s post-hoc test, with statistical significance set at P < 0.05.
Data availability
All relevant data for this study are included within the manuscript. The raw data that support the findings of this study are available from the corresponding author upon reasonable request.
References
WORLD HEALTH ORGANIZATION & Schistosomiasis (2024). https://www.who.int/publications/i/item/9789241503129
WORLD HEALTH ORGANIZATION. Ending the neglect to attain the Sustainable Development Goals: a road map for neglected tropical diseases 2021–2030. (2021). https://www.who.int/publications/i/item/9789240010352
de Moraes, J. & Geary, T. G. FDA-approved antiparasitic drugs in the 21st Century: a success for helminthiasis? Trends Parasitol. 36, 573–575. https://doi.org/10.1016/j.pt.2020.04.005 (2020).
McManus, D. P. et al. Nat. Rev. Dis. Primers 4, 13 doi: https://doi.org/10.1038/s41572-018-0013-8. (2018).
Mengarda, A. C., Iles, B., Longo, J. P. & de Moraes, J. Recent trends in praziquantel nanoformulations for helminthiasis treatment. Expert Opin. Drug Deliv. 19, 383–393. https://doi.org/10.1080/17425247.2022.2051477 (2022).
Spangenberg, T. Alternatives to praziquantel for the prevention and control of schistosomiasis. ACS Infect. Dis. 7, 939–942. https://doi.org/10.1021/acsinfecdis.0c00542 (2021).
Ferreira, L. L. G., de Moraes, J. & Andricopulo, A. D. Approaches to advance drug discovery for neglected tropical diseases. Drug Discov Today. 27, 2278–2287. https://doi.org/10.1016/j.drudis.2022.04.004 (2022).
de Wit, L. A. & Ricketts, T. H. Trade and Deforestation predict rat lungworm disease, an invasive-driven zoonosis, at global and regional scales. Front. Public. Health. 9, 680986. https://doi.org/10.3389/fpubh.2021.680986 (2021).
Cowie, R. H. Angiostrongylus cantonensis: Agent of a sometimes fatal globally emerging infectious disease (rat lungworm disease). ACS Chem. Neurosci. 8, 2102–2104. https://doi.org/10.1021/acschemneuro.7b00335 (2017).
Federspiel, F., Skovmand, S. & Skarphedinsson, S. Eosinophilic meningitis due to Angiostrongylus cantonensis in Europe. Int. J. Infect. Dis. 93, 28–39. https://doi.org/10.1016/j.ijid.2020.01.012 (2020).
Roquini, D. B. et al. Susceptibility of Angiostrongylus cantonensis larvae to anthelmintic drugs. Front. Pharmacol. 21, 13, 901459. https://doi.org/10.3389/fphar.2022.901459 (2022).
Roquini, D. B. et al. Antihistamines H1 as potential anthelmintic agents against the zoonotic parasite Angiostrongylus cantonensis. ACS Omega. 9, 31159–31165. https://doi.org/10.1021/acsomega.4c04773 (2024).
Dube, N. P. et al. Review on the applications of selected metal-based complexes on infectious diseases. Molecules 29, 406. https://doi.org/10.3390/molecules29020406 (2024).
Anthony, E. J. et al. Metallodrugs are unique: opportunities and challenges of discovery and development. Chem. Sci. 11, 12888–12917. https://doi.org/10.1039/d0sc04082g (2020).
Tóth, B., Hohmann, J., Vasas, A. & Phenanthrenes A promising group of plant secondary metabolites. J. Nat. Prod. 81, 661–678. https://doi.org/10.1021/acs.jnatprod.7b00619 (2018).
Barta, A. et al. Phenanthrenes from Juncus articulatus with antibacterial and biofilm formation inhibitory activity. J. Nat. Prod. 87, 2068–2080. https://doi.org/10.1021/acs.jnatprod.4c00577 (2024).
Deegan, C., Coyle, B., McCann, M., Devereux, M. & Egan, D. A. Vitro vitro anti-tumour effect of 1,10-phenanthroline-5,6-dione (phendione), [Cu(phendione)3](ClO4)2.4H2O and [Ag(phendione)2]ClO4 using human epithelial cell lines. Chem. Biol. Interact. 164, 115–125. https://doi.org/10.1016/j.cbi.2006.08.025 (2006).
Lenis-Rojas, O. A. et al. In vitro and in vivo biological activity of ruthenium 1,10-phenanthroline-5,6-dione arene complexes. Int. J. Mol. Sci. 23, 13594 (2022). https://doi.org/10.3390/ijms232113594
Viganor, L. et al. Anti-Pseudomonas aeruginosa activity of 1,10-phenanthroline-based drugs against both planktonic- and biofilm-growing cells. J. Antimicrob. Chemother. 71, 128–134. https://doi.org/10.1093/jac/dkv292 (2016).
Choroba, K. et al. In vitro and in vivo biological activities of dipicolinate oxovanadium(IV) Complexes. J. Med. Chem. 66, 8580–8599 (2023). https://doi.org/10.1021/acs.jmedchem.3c00255
Vargas Rigo, G. et al. Anti-Trichomonas vaginalis activity of 1,10-phenanthroline-5,6-dione-based metallodrugs and synergistic effect with metronidazole. Parasitology 146, 1179–1183. https://doi.org/10.1017/S003118201800152X (2019).
Oliveira, S. S. C. et al. The Anti-Leishmania amazonensis and Anti-Leishmania Chagasi action of copper(II) and silver(I) 1,10-phenanthroline-5,6-dione coordination compounds. Pathogens 12, 70. https://doi.org/10.3390/pathogens12010070 (2023).
Lima, A. K. C. et al. Anti-Leishmania braziliensis activity of 1,10-phenanthroline-5,6-dione and its Cu(II) and ag(I) complexes. Parasitol. Res. 120, 3273–3285. https://doi.org/10.1007/s00436-021-07265-x (2021).
de Moraes, J., Dario, B. S., Couto, R. A. & Pinto, P. L. Da Costa Ferreira, A.M. Antischistosomal activity of oxindolimine-metal complexes. Antimicrob. Agents Chemother. 59, 6648–6652. https://doi.org/10.1128/AAC.01371-15 (2015).
Rigo, G. V. et al. Peptidases are potential targets of copper(II)-1,10-phenanthroline-5,6-dione complex, a promising and potent new drug against Trichomonas vaginalis. Pathogens 12, 745. https://doi.org/10.3390/pathogens12050745 (2023).
Gu, C. et al. Ivermectin induces oxidative stress and mitochondrial damage in Haemonchus Contortus. Vet. Parasitol. 333, 110352. https://doi.org/10.1016/j.vetpar.2024.110352 (2025).
Nagdy, Y. A. E., Nabil, Z. E., El-Shenawy, N. S. & Elkhawass, E. A. Antiparasitic and antioxidant effects of selenium nanoparticles on parasitic trichinella spiralis. Exp. Parasitol. 268, 108876. https://doi.org/10.1016/j.exppara.2024 (2025).
Brito, J. R. et al. Neolignans isolated from Saururus cernuus L. (Saururaceae) exhibit efficacy against Schistosoma mansoni. Sci. Rep. 12, 19320. https://doi.org/10.1038/s41598-022-23110-2 (2022).
Sessa, D. P. et al. 15β-Senecioyl-oxy-ent-kaur-16-en-19-oic Acid, a diterpene isolated from Baccharis Lateralis, as promising oral compound for the treatment of schistosomiasis. J. Nat. Prod. 83, 3744–3750. https://doi.org/10.1021/acs.jnatprod.0c01050 (2020).
Mengarda, A. C. et al. Licarin A, a neolignan isolated from Nectandra oppositifolia Nees & Mart. (Lauraceae), exhibited moderate preclinical efficacy against Schistosoma mansoni infection. Phytother Res. 35, 5154–5162 (2021). https://doi.org/10.1002/ptr.7184
Rocha, V. C. et al. Evaluating the antischistosomal activity of dehydrodieugenol B and its methyl ether isolated from Nectandra leucantha - A preclinical study against Schistosoma mansoni infection. ACS Omega. 8, 40890–40897. https://doi.org/10.1021/acsomega.3c06111 (2023).
Silva, B. C. et al. Efficacy of carvacryl acetate in vitro and following oral administration to mice harboring either prepatent or patent Schistosoma mansoni infections. Parasitol. Res. 120, 3837–3844. https://doi.org/10.1007/s00436-021-07333-2 (2021).
Queiroz, L. S. et al. In vitro and in vivo evaluation of cnicin from blessed thistle (Centaurea benedicta) and its inclusion complexes with cyclodextrins against Schistosoma mansoni. Parasitol. Res. 120, 1321–1333 (2021). https://doi.org/10.1007/s00436-020-06963-2
Silva, L. M. et al. Licochalcone A-loaded solid lipid nanoparticles improve antischistosomal activity in vitro and in vivo. Nanomedicine 16, 1641–1655. https://doi.org/10.2217/nnm-2021-0146 (2021).
D Costa, P. et al. Assessment of the in vitro antischistosomal activities of the extracts and compounds from Solidago Microglossa DC (Asteraceae) and Aristolochia Cymbifera Mart. & Zucc. (Aristolochiaceae). Evid. Based Complement. Alternat Med. 2020 (1726365). https://doi.org/10.1155/2020/1726365 (2020).
Xavier, E. S. et al. Therapeutic efficacy of carvacrol-loaded nanoemulsion in a mouse model of schistosomiasis. Front. Pharmacol. 13, 917363. https://doi.org/10.3389/fphar.2022.917363 (2022).
Albuquerque, M. M. S. et al. Oral administration of kaempferol isolated from Baccharis mattogrosensis enables in vivo activity against Schistosoma mansoni. Chem. Biodivers. e202401452 (2024). (2024). https://doi.org/10.1002/cbdv.202401452
McCann, M. et al. In vitro and in vivo studies into the biological activities of 1,10-phenanthroline, 1,10-phenanthroline-5,6-dione and its copper(II) and silver(I) complexes. Toxicol. Res. 1, 47–54. https://doi.org/10.1039/c2tx00010e (2012).
Gandra, R. M. et al. In vivo activity of copper(II), manganese(II), and silver(I) 1,10-phenanthroline chelates against Candida haemulonii using the Galleria mellonella model. Front Microbiol. 11, 470 (2020). https://doi.org/10.3389/fmicb.2020.00470
Santos, A. L. S. et al. Decoding the anti-Leishmania braziliensis activity of 1,10-phenanthroline-5,6-dione and its silver- and copper-based complexes: In vitro and in vivo approaches. Eur. J. Med. Chem. Rep. 6, 100093 (2022). https://doi.org/10.1016/j.ejmcr.2022.100093
Vaidya, S. P., Gadre, S., Kamisetti, R. T. & Patra, M. Challenges and opportunities in the development of metal-based anticancer theranostic agents. Biosci. Rep. 42, BSR20212160. https://doi.org/10.1042/BSR20212160 (2022).
Boros, E., Dyson, P. J. & Gasser, G. Classification of metal-based drugs according to their mechanisms of. Action Chem. 6, 41–60. https://doi.org/10.1016/j.chempr.2019.10.013 (2020).
McCann, M. et al. Synthesis and X-ray crystal structure of [Ag(phendio)2]ClO4 (phendio = 1,10-phenanthroline- 5,6-dione) and its effects on fungal and mammalian cells. Biometals 17, 635–645. https://doi.org/10.1007/s10534-004-1229-5 (2004).
Roquini, D. B. et al. Promethazine exhibits antiparasitic properties in vitro and reduces worm burden, egg production, hepato-, and splenomegaly in a schistosomiasis animal model. Antimicrob. Agents Chemother. 63, e01208–e01219. https://doi.org/10.1128/AAC.01208-19 (2019).
Xavier, R. P. et al. H1-antihistamines as antischistosomal drugs: in vitro and in vivo studies. Parasit. Vectors. 13, 278. https://doi.org/10.1186/s13071-020-04140-z (2020).
Silva, T. C. et al. New evidence for tamoxifen as an antischistosomal agent: in vitro, in vivo and target fishing studies. Future Med. Chem. 13, 945–957. https://doi.org/10.4155/fmc-2020-0311 (2021).
Brenner, S. The genetics of Caenorhabditis elegans. Genetics 77, 71–94. https://doi.org/10.1093/genetics/77.1.71 (1974).
Souza, D. C. S. et al. In vivo antischistosomal efficacy of PorPonderosaderosa γ-lactones. Phytomedicine 135, 156045. https://doi.org/10.1016/j.phymed.2024.156045 (2024).
Cai, H. et al. Systemic toxicity evaluation of novel tobacco products in Caenorhabditis elegans. Toxicol. Vitro. 62, 104671. https://doi.org/10.1016/j.tiv.2019.104671 (2020).
Pavani, T. F. A., Cirino, M. E., Teixeira, T. R., de Moraes, J. & Rando, D. G. G. Targeting the Schistosoma mansoni nutritional mechanisms to design new antischistosomal compounds. Sci. Rep. 13, 19735. https://doi.org/10.1038/s41598-023-46959-3 (2023).
Amorim, C. R. et al. Schiff bases of 4-phenyl-2-aminothiazoles as hits to new antischistosomals: Synthesis, in vitro, in vivo and in silico studies. Eur. J. Pharm. Sci. 150, 105371 (2020). https://doi.org/10.1016/j.ejps.2020.105371
Silva, T. C. et al. N-(4-Methoxyphenyl)Pentanamide, a simplified derivative of albendazole, displays anthelmintic properties against the nematode Toxocara canis. Microbiol. Spectr. 10, e0180722. https://doi.org/10.1128/spectrum.01807-22 (2022).
Moreira-Filho, J. T., Neves, B. J., Cajas, R. A., Moraes, J. & Andrade, C. H. Artificial intelligence-guided approach for efficient virtual screening of hits against Schistosoma mansoni. Future Med. Chem. 15, 2033–2050. https://doi.org/10.4155/fmc-2023-0152 (2023).
Morais, C. S. et al. Pyrazoline derivatives as promising novel antischistosomal agents. Sci. Rep. 11, 23437. https://doi.org/10.1038/s41598-021-02792-0 (2021).
Acknowledgements
This work was supported by the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, Grant 2023/08418-6). MEC, TRT, and LFS received postgraduate fellowships from the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, Finance Code 001). ACCB was awarded a fellowship from the Institutional Scientific Initiation Scholarship Program of the Conselho Nacional de Desenvolvimento Científico e Tecnológico (PIBIC-CNPq). JdM and A.L.S.S. hold Research Productivity Grants from CNPq. The funding agencies had no involvement in the study’s design, data collection, analysis, interpretation, or the preparation of this manuscript. The authors also thank Bruna L. Lemes and Daniel B. Roquini for their assistance with the A. cantonensis experiments. Special thanks to Mariana (B) Silva for her technical support at the Núcleo de Pesquisa em Doenças Negligenciadas, Guarulhos University, Brazil.
Author information
Authors and Affiliations
Contributions
M.E.C., A.L.S.S., and J.d.M. contributed to conceptualization; M.E.C., T.R.T., A.C.C.B., L.F.S., and A.M.H.S., contributed to experiments and data interpretation; M.E.C. and J.d.M. contributed to writing - original draft and visualization; A.M.H.S., A.L.S.S., and J.d.M. contributed with resources, writing - review & editing; J.d.M. contributed to project administration and funding acquisition. All authors have read and agreed to the published version of the manuscript.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethical approval
This study adhered to the ARRIVE guidelines established by the National Centre for the Replacement, Refinement & Reduction of Animals in Research (NC3Rs). All experimental protocols were approved by the Committee for the Ethical Use of Animals in Experimentation at Guarulhos University (Guarulhos, SP, Brazil), under protocol numbers 064/24 (A. cantonensis) and 065/24 (S. mansoni). The study was conducted in full compliance with Brazilian legislation on the care and use of laboratory animals.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Cirino, M.E., Teixeira, T.R., Silva, A.M.H. et al. Anthelmintic activity of 1,10-phenanthroline-5,6-dione-based metallodrugs. Sci Rep 15, 4699 (2025). https://doi.org/10.1038/s41598-025-88484-5
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41598-025-88484-5