Next Article in Journal
Antibiotic Residues and Resistance in Three Wastewater Treatment Plants in Romania
Previous Article in Journal
Farnesol Emulsion as an Effective Broad-Spectrum Agent against ESKAPE Biofilms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

EDBD—3,6-Epidioxy-1,10-Bisaboladiene—An Endoperoxide Sesquiterpene Obtained from Drimys brasiliensis (Winteraceae) Exhibited Potent Preclinical Efficacy against Schistosoma mansoni Infection

by
Eric Umehara
1,
Thainá R. Teixeira
2,
Rayssa A. Cajás
2,
Monique C. Amaro
2,
Josué de Moraes
2,3,* and
João Henrique G. Lago
1,*
1
Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André 09280-560, SP, Brazil
2
Centro de Pesquisas de Doenças Negligenciadas, Universidade Guarulhos, Guarulhos 07023-070, SP, Brazil
3
Núcleo de Pesquisas em Doenças Negligenciadas, Instituto Científico e Tecnológico, Universidade Brasil, São Paulo 08230-030, SP, Brazil
*
Authors to whom correspondence should be addressed.
Antibiotics 2024, 13(8), 779; https://doi.org/10.3390/antibiotics13080779
Submission received: 17 June 2024 / Revised: 7 August 2024 / Accepted: 15 August 2024 / Published: 18 August 2024
(This article belongs to the Special Issue Antiparasitic Natural Products)

Abstract

:
Schistosomiasis, a neglected tropical disease impacting over 250 million individuals globally, remains a major public health challenge due to its prevalence and significant impact on affected communities. Praziquantel, the sole available treatment, highlights the urgency of the need for novel anthelmintic agents to achieve the World Health Organization (WHO) goal of schistosomiasis elimination. Previous studies reported the promising antiparasitic activity of different terpenoids against Schistosoma mansoni Sambon (Diplostomida: Schistosomatidae). In the present work, the hexane extract from branches of Drimys brasiliensis afforded a diastereomeric mixture of endoperoxide sesquiterpenes, including 3,6-epidioxy-bisabola-1,10-diene (EDBD). This compound was evaluated in vitro and in vivo against S. mansoni. EDBD exhibited a significant reduction in S. mansoni viability in vitro, with an effective concentration (EC50) value of 4.1 µM. Additionally, EDBD demonstrated no toxicity to mammalian cells. In silico analysis predicted good drug-likeness properties, adhering to pharmaceutical industry standards, including favorable ADME profiles. Furthermore, oral treatment of S. mansoni-infected mice with EDBD (400 mg/kg) resulted in a remarkable egg burden reduction (98% and 99% in tissues and feces, respectively) surpassing praziquantel’s efficacy. These findings suggest the promising potential of EDBD as a lead molecule for developing a novel schistosomiasis treatment.

1. Introduction

Worm parasites contribute significantly to the global disease burden, affecting nearly 1.8 billion people worldwide, particularly in regions with inadequate healthcare and socioeconomic conditions. Among these parasites, the blood fluke of the genus Schistosoma is noteworthy for its responsibility in nearly 240 million cases of schistosomiasis worldwide, posing a risk of infection to approximately 10% of the human population [1]. Chronic infection can lead to significant morbidity, with an estimated 1.5 million disability-adjusted life years (DALYs) lost due to schistosomiasis [2]. Schistosoma mansoni Sambon (Diplostomida: Schistosomatidae) adult worms reside within the human portal vasculature, often for years. They release eggs that can travel in two paths: either passing through the intestinal wall and exiting in the feces or migrating to the liver. In the liver, the eggs trigger an immune response, leading to the formation of granulomas and peri-portal fibrosis [3].
The World Health Organization’s (WHO) neglected tropical diseases roadmap 2021–2030 aims to eliminate schistosomiasis as a public health issue and interrupt its transmission in selected countries [4]. However, the current strategy heavily relies on praziquantel (PZQ), leading to concerns about drug resistance and efficacy due to its widespread use [5]. Given the limitations of PZQ, there is a pressing need for alternative treatments [6]. Natural products are pivotal in drug discovery for treating infectious diseases due to their unique characteristics compared to synthetic molecules [7]. Plants of the genus Drimys (Winteraceae), including D. brasiliensis, are rich sources of specialized metabolites, such as sesquiterpenes with antifungal [8], anti-Leishmania [9], antimalarial [9], and anti-inflammatory activity [10] as well as alkaloids with antinociceptive effects [11]. D. brasiliensis, found across the Brazilian Atlantic Forest, Cerrado, and Caatinga biomes, holds particular promise in this regard [12]. While previous research has explored sesquiterpenes from D. brasiliensis for anti-Trypanosoma cruzi activity [13], their potential against Schistosoma remains unexplored.
The aim of this study was to evaluate the antiparasitic properties of 3,6-epidioxy-1,10-bisaboladiene (EDBD), a diastereomeric mixture of endoperoxide sesquiterpenes obtained from D. brasiliensis, against Schistosoma mansoni both in vitro and in an animal model of schistosomiasis. In vitro experiments determined its effective concentration 50% (EC50), while in vivo studies assessed its efficacy in S. mansoni-infected mice. Additionally, the toxicity profile of EDBD was evaluated in mammalian cells, providing valuable insights into its safety and potential efficacy.

2. Results

2.1. Chemical Characterization of EDBD

ESI-HRMS (Figure S1) showed a [M+H]+ ion peak at m/z 237.1852 (calculated for C15H25O2, 237.1854), confirming the molecular formula as C15H24O2, with four unsaturation. 1H NMR spectrum (Figure S2) displayed, along with other signals, two coupled doublets (J = 8.5 Hz) at δ 6.45 and 6.38, assigned to the olefinic hydrogens H-1 and H-2, respectively, whereas the triplet at δ 5.08 (J = 6.0 Hz) was attributed to H-10. The presence of four methyl groups was inferred by the occurrence of three singlets at δ 1.67 (H-12), 1.60 (H-13), and 1.37 (H-15) and one doublet at δ 0.99 (J = 6.4 Hz, H-14).
The 13C (Figure S3) and DEPT (Figure S4) NMR spectra showed 15 signals, being four methyl, four methylene, four methine and three quaternaries, confirming the occurrence of a sesquiterpene. Among these signals, four corresponding to sp2 carbons were observed at δ 136.3 (C-1), 133.5 (C-2), 124.1 (C-10) and 131.8 (C-11). Additionally, two carbinolic carbons were observed at δ 79.9 (C-6) and 74.4 (C-3), suggesting the presence of an endoperoxide moiety.
Finally, analysis of HSQC (Figure S5) and HMBC (Figure S6) spectra followed by comparison of the obtained data with those previously published [14], confirmed the chemical structure of 3,6-epidioxy-bisabola-1,10-diene—EDBD (Figure 1).

2.2. EDBD Exhibited Antiparasitic Effects on S. mansoni without Toxicity to Mammalian Cells

The in vitro antiparasitic effect of EDBD against S. mansoni adult worms (both male and female) was evaluated to determine the effective concentration 50% (EC50). PZQ served as the positive while 0.5% DMSO, representing the highest concentration of solvent, was used as the negative control. As shown in Figure 2, parasites in the negative control group (0.5% DMSO) remained viable throughout the incubation period. In contrast, PZQ caused the mortality of all schistosomes immediately. EDBD exhibited significant antiparasitic effects at low micromolar concentrations. The EC50 values are summarized in Table 1. EDBD displayed marked anthelmintic activity, with EC50 values of 4.1 μM for both male and female adult schistosomes. These values are comparable to the positive control PZQ, which had EC50 values of 1.1 μM and 1.3 μM, respectively.
To evaluate toxicity, the Vero mammalian cell line was used to determine the CC50 and the selectivity index (SI) values of EDBD. Notably, EDBD exhibited no cytotoxicity against Vero cells (Figure 2), with CC50 values exceeding 200 μM, resulting in a favorable SI > 48 for adult schistosomes (Table 1), underscoring the compound’s safety profile in an animal model.

2.3. In Silico Evaluation

Considering the in vitro activity of EDBD, an in silico analysis was conducted using the SwissADME platform, which investigates the pharmacokinetic and pharmacodynamic properties of molecular structures. An important tool for interpreting these data is the bioavailability radar (Figure 3), which assesses a drug’s absorption rate into the bloodstream and its biotransformation for excretion. Upon reviewing the image, it is observed that DBD exhibits properties similar to those of the positive control, PZQ.
Table 2 presents the in silico physicochemical values and ADME parameters of EDBD in comparison to PZQ. Notably, EDBD does not violate any of Lipinski’s, Ghose’s, Veber’s, Egan’s, or Muegge’s rules, indicating they have a molecular weight (MW) < 500 Da, log Po/w < 5, and topological polar surface area (TPSA) < 140 Å. These values suggest high absorption and good oral availability [15], positioning them as lead-like compounds. Additionally, no alerts for PAINS were detected for either tested compound.

2.4. In Vivo Evaluation of EDBD—Reduction in Adult Worm Parasite Burden and a Decrease in Egg Production

Based on the promising in vitro and in silico findings, in vivo testing of EDBD was conducted in S. mansoni-infected mice. Animals were orally administered 400 mg/kg of EDBD, the standard dose used in schistosomiasis drug discovery programs with a murine model infected with S. mansoni [16], and compared with infected but untreated controls. Treatment with EDBD led to a significant reduction in parasite burden by 61.20%, while the antiparasitic drug PZQ caused a worm burden reduction of 85.03% (Figure 4).
In addition to parasite burden reduction, treatment with EDBD resulted in a marked decrease in egg production, with reductions of 99.22% in feces and 98.18% in intestines (p < 0.001), surpassing the effectiveness of PZQ, which achieved reductions of 84% to 89% in egg burden (Figure 5).

3. Discussion

In this study, the diastereomeric mixture (1:1) of endoperoxide sesquiterpene 3,6-epidioxy-bisabola-1,10-diene (EDBD) was obtained from the hexane extract of D. brasiliensis branches and was characterized by NMR (¹H and ¹³C) and ESI-HRMS analysis. The biological results of this study demonstrate that EDBD exhibits significant antiparasitic activity against S. mansoni, a major parasitic pathogen causing schistosomiasis. The effectiveness of EDBD is underscored by its reduced EC50 value (4.1 μM) for both male and female adult worms, which is comparable to the commonly used antiparasitic drug PZQ. This highlights the potential of EDBD as a viable alternative or complement to existing treatments.
The significance of the endoperoxide group is particularly notable, as literature data describe its importance for the antiparasitic activity of similar molecules such as ascaridole and artemisinin [17]. Although the reasons for sex-dependent drug sensitivity in S. mansoni are not fully understood, several natural products with antischistosomal properties are known to exert varying effects on male and female parasites [18]. It is possible that the targets of EDBD are present in both sexes.
Evaluating the safety profiles of compounds is critical in the antischistosomal drug discovery process to prioritize chemical entities devoid of overt toxicity [18,19]. In this study, the in vitro assays indicated that EDBD effectively targets adult schistosomes without exhibiting cytotoxicity toward Vero mammalian cells. The high selectivity index (SI > 48) further emphasizes its safety and efficacy, distinguishing it from other compounds that may show toxicity at therapeutic doses [20].
The in silico analysis provided insights into the pharmacokinetic and pharmacodynamic properties of EDBD in comparison to the control PZQ. EDBD exhibited excellent compliance with several drug-likeness rules, which predict high absorption and oral bioavailability. These properties are critical for the development of oral medications [21], suggesting that EDBD is a promising candidate for further drug development.
The in vivo studies further validated the antiparasitic potential of EDBD. In S. mansoni-infected mice, oral administration of the compound significantly reduced the parasite burden by 61.20%. Although this reduction is substantial and demonstrates EDBD’s ability to combat schistosome infections in a living organism, PZQ showed superior efficacy, achieving an 85.03% reduction in worm burden. This higher efficacy may be attributed to the rapid action of PZQ in killing adult worms. Notably, EDBD led to an almost complete reduction in egg production, with reductions of 99.22% in feces and 98.18% in intestines, surpassing the efficacy of PZQ in this regard. Since egg production is a major factor in the pathology and transmission of schistosomiasis [4], the ability of EDBD to inhibit egg production so effectively is particularly noteworthy.
For comparison, previous studies have reported varying levels of efficacy with different compounds, including several natural products. The lignan licarin A, at a single oral dose of 400 mg/kg, showed a 46.25% reduction in adult worms [22], while the sesquiterpene nerolidol exhibited a 70.1% reduction in adult worms and a 69.7% reduction in eggs within the intestine [23]. Notably, a single oral dose of 400 mg/kg of EDBD effectively impacted S. mansoni-infected mice more than these compounds reported in the literature [22,23], both in terms of adult worm reduction and egg deposition. These findings suggest that EDBD could serve as a promising prototype for schistosomiasis treatment. Additionally, they open up the possibility for future studies on the synergistic effects of these compounds and their structure–activity relationships with similar molecules, such as ascaridole. Further research could explore these aspects to optimize the antiparasitic efficacy and safety of EDBD and related compounds.
Finally, several limitations should be acknowledged in this study. Firstly, the pharmacokinetic properties of EDBD were predicted in silico, and no in vivo pharmacokinetic studies were performed. Consequently, the actual ADME profile of EDBD remains unknown, which could impact its effectiveness and safety in a clinical setting. Additionally, this study focused solely on a murine model, which may not fully capture the drug’s efficacy and safety in humans. Future research should include comprehensive pharmacokinetic evaluations and clinical trials to confirm EDBD’s potential as an effective treatment for schistosomiasis.

4. Materials and Methods

4.1. General Procedures

NMR spectra were recorded using CDCl₃ (Sigma-Aldrich, St Louis, MI, USA) as the solvent and TMS as the internal standard on a Varian INOVA spectrometer, operating at 500 MHz for 1H and 125 MHz for 13C nuclei. The ESI-HRMS spectrum was measured on a Bruker Daltonics MicroTOF QII spectrometer. For column chromatography (C.C), silica gel (230−400 mesh, Merck, Darmstadt, Germany) was used, while silica gel 60 PF254 (Merck, Darmstadt, Germany) was employed for analytical TLC separations.

4.2. Plant Material

Branches of D. brasiliensis were collected in March 2021 from the Serra do Cipó National Park, Minas Gerais State, Brazil, by Dr. Guilherme M. Antar. The collection was registered under code A4123E4 in SISGEN. A voucher specimen (SPF4105) has been archived at the Botanical Institute of São Paulo, São Paulo State, Brazil.

4.3. Isolation of EDBD (3,6-Epidioxy-Bisabola-1,10-Diene)

Dried branches of D. brasiliensis (316 g) were exhaustively extracted using hexane (10 × 500 mL) at room temperature. After evaporation of the solvent under reduced pressure, 16 g of crude extract was obtained. A portion of this extract (15 g) was subjected to chromatography on a silica gel column eluted with increasing amounts of hexane/ethyl acetate (1:0, 9:1, 8:2, 7:3, 6:4, 3:7, and 0:1), resulting in 21 fractions that were combined into five groups (A–E). Part of group C (300 mg) was further purified by silica gel column chromatography using hexane/ethyl ether (98:2, 9:2, 8:2, and 1:1) to yield 140 mg of EDBD.
EDBD (3,6-epidioxy-bisabola-1,10-diene), pale yellow oil. 1H NMR (500 MHz, CDCl3): δ 6.45 (d, J = 8.5 Hz, H-1), 6.38 (d, J = 8.5 Hz, H-2), 5.08 (t, J = 6.0 Hz, H-10), 2.10 (m, H-9a), 2.02 (m, H-4a), 1.94 (m, H-9b), 1.93 (m, H-5a), 1.73 (m, H-7) 1.67 (br s, H-12), 1.62 (m, H-8a), 1.60 (br s, H-13), 1.50 (m, H-4b), 1.37 (s, H-15), 1.14 (m, H-8b) 0.99 (d, J = 6.4 Hz, H-14); 13C NMR (125 MHz, CDCl3): δ 136.3 (C-2), 133.5 (C-1), 131.8 (C-11) 124.1 (C-10), 79.9 (C-6), 74.4 (C-3) 36.7 (C-7) 31.1 (C-8), 29.4 (C-4) 26.0 (C-9), 25.8 (C-12) 25.5 (C-5) 21.4 (C-15), 17.6 (C-13) 13.8 (C-14); ESI-HRMS m/z 237.1852 [M + H]+ (calculated for C15H25O2 237.1854).

4.4. In Silico Analysis

The pharmacokinetics, drug likeness, and medicinal chemistry parameters of EDBD (3,6-epidioxy-1,10-bisaboladiene) and PZQ were evaluated in silico using the SwissADME platform (http://www.swissadme.ch/, accessed on 1 June 2024) [24]. Several parameters were evaluated: (i) Absorption, Distribution, Metabolism, and Excretion (ADME); (ii) physicochemical properties such as the number of rotatable bonds, H-bond donors, and H-bond acceptors; (iii) lipophilicity (log p value); (iv) pharmacokinetics aspects like gastrointestinal absorption and CYP 450 inhibition; (v) drug likeness criteria based on the Lipinski (Pfizer, Hong Kong), Veber (GlaxoSmithKline, Hong Kong), and Muegge (Bayer, Leverkusen, Germany) filters; and (vi) identification of pan-assay interference compounds (PAINS).

4.5. Animals, Worms and Cells

The life cycle of the helminths (Belo Horizonte [BH] strain) was sustained through passage in Biomphalaria glabrata snails and Swiss mice at the Research Center on Neglected Diseases, Guarulhos University, SP, Brazil. The snails and mice were kept under controlled environmental conditions (25 °C, 50% humidity) with unrestricted access to food and water. Vero cells obtained from the American Type Culture Collection (ATCC) were cultured in DMEM supplemented with 10% fetal bovine serum (FBS) and 2 mM L-glutamine [25].

4.6. In Vitro Antischistosomal Assay

The in vitro anthelmintic assay followed established protocols [26]. Adult schistosomes, obtained from infected mice, were exposed to varying concentrations (0.78 to 50.0 μM) of EDBD and PZQ in RPMI 1640 culture medium supplemented with 5% heat-inactivated fetal calf serum, 100 U/mL penicillin, and 100 μg/mL streptomycin in 24-well culture plates (Sigma-Aldrich, St Louis, MI, USA). Each well contained one pair of parasites (one male and one female). The compounds were dissolved in 0.5% DMSO, and each concentration was tested in triplicate, with the experiments repeated three times for reliability.
Parasite motility was observed daily at 24, 48, and 72 h using a BEL Engineering microscope (INV 100, Monza [MB], Italy). A worm was considered deceased if it remained motionless everywhere on its body for at least 1 min, even upon stimulation with tweezers.

4.7. Toxicity Profile in Mammalian Cells

The cytotoxicity assessment followed established protocols [21,22]. Cells were cultured in 96-well plates using DMEM supplemented with 10% heat-inactivated fetal calf serum and 2 mM L-glutamine. After allowing 24 h for cell adhesion at 37 °C and 5% CO2, the 50% cytotoxic concentrations (CC50) of EDBD and PZQ were determined over a concentration range of 0.12–200 μM. Following a 72-h incubation period, MTT solution was added, and the plates were further incubated for 3 h. Subsequently, the absorbance was measured at 595 nm using an Epoch Microplate Spectrophotometer (BioTek Instruments, Winooski, VT, USA). The assay was performed in triplicate and repeated three times to ensure reliability. Results were calculated as a percentage relative to the control.
Selectivity indices (SI) were calculated by dividing the CC50 values obtained in Vero cells by those determined in S. mansoni.

4.8. In Vivo Therapeutic Efficacy in an Animal Model of Schistosomiasis

Female mice (3 weeks old) were subcutaneously infected with 80 S. mansoni cercariae each. At forty-two days post-infection, corresponding to the presence of adult worms (patent infection), EDBD was administered as a single dose of 400 mg/kg to a group of five mice [27,28]. Control groups included S. mansoni-infected mice treated with a vehicle or PZQ at 400 mg/kg (five mice per group).
At 56 days post-infection, all experimental groups were weighed, euthanized, and dissected. Schistosomes were collected from the hepatic portal system and mesenteric veins, and their numbers were counted after sexing. Therapeutic efficacy was assessed qualitatively and quantitatively by examining the presence of immature eggs in the intestine. Additionally, the Kato–Katz method was used for quantitative fecal examination to provide further insights into the treatment outcomes [29].

4.9. Statistical Analysis

Data were analyzed using GraphPad Prism 8.0. CE50 and CC50 values were obtained from the dose–response curves. The Kruskal–Wallis test was used for in vivo studies, with a p value of <0.05 considered statistically significant.

5. Conclusions

In conclusion, the results of the present study demonstrated the anti-schistosomal activity of a diastereomeric mixture of the endoperoxide sesquiterpene 3,6-epidioxy-1,10-bisaboladiene (EDBD) obtained from branches of D. brasiliensis (Winteraceae). In vitro assays showed worm mortality with EC50 values of 4.1 μM, reducing the number of male and female parasites. Based on these in vitro results, EDBD underwent in vivo assays using a murine infection model, where there was a reduction in worm burden (61.20%) and in egg production in feces (99.22%) and tissues (98.18%), indicating superior efficacy of the compound compared to the positive control PZQ. Importantly, EDBD did not demonstrate cytotoxicity in mammalian cells. Therefore, EDBD represents a promising candidate for the development of new therapeutic approaches against schistosomiasis.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antibiotics13080779/s1, Figure S1: ESI-HR spectrum (positive mode) spectrum of EDBD—3,6-epidioxy-1,10-bisaboladiene, Figure S2: 1H NMR spectrum of EDBD—3,6-epidioxy-1,10-bisaboladiene, Figure S3: 13C NMR spectrum of EDBD—3,6-epidioxy-1,10-bisaboladiene, Figure S4: DEPT NMR spectrum of EDBD—3,6-epidioxy-1,10-bisaboladiene, Figure S5: HSQC spectrum of EDBD—3,6-epidioxy-1,10-bisaboladiene, Figure S6: HMBC spectrum of EDBD—3,6-epidioxy-1,10-bisaboladiene.

Author Contributions

E.U., T.R.T., R.A.C. and M.C.A., data curation, visualization, investigation, writing original draft preparation; J.d.M. and J.H.G.L., conceptualization, methodology, writing, reviewing, and editing the final version. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Fundação de Amparo à Pesquisa do Estado de São Paulo—FAPESP (grants 23/08418-6 and 23/12447-1). J.deM. and J.H.G.L. also received the Established Investigator Fellowship from Conselho Nacional de Desenvolvimento Científico e Tecnológico—CNPq (Brazil). E.U., R.A.C. and T.R.T. received a post-graduate fellowship from the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—CAPES (Finance Code 001, Brazil). This publication is part of the activities of the Research Network Natural Products against Neglected Diseases (ResNetNPND): http://www.uni-muenster.de/ResNetNPND/, accessed on 1 June 2024.

Institutional Review Board Statement

The animal studies are reported in compliance with the National Centre for the Replacement and Refinement & Reduction of Animals in Research (NC3Rs) ARRIVE guidelines. All experiments were conducted in conformity with the Brazilian law for Guidelines for Care and Use of Laboratory Animals [Law 11790/2008]. The protocol for the experimental design was approved by the Comissão de Ética no Uso de Animais (CEUA) at Universidade Guarulhos (São Paulo, Brazil—protocol ID 47/20 on June 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Acknowledgments

The authors thank the multiuser central facilities (UFABC) for the experimental support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. World Health Organization. Schistosomiasis; World Health Organization: Geneva, Switzerland, 2023; Available online: https://www.who.int/news-room/fact-sheets/detail/schistosomiasis (accessed on 16 April 2024).
  2. Anissuzaman; Tsuji, N. Schistosomiasis and hookworm infection in humans: Disease burden, pathology and anthelmintic vaccines. Parasitol. Int. 2020, 75, 102051. [Google Scholar] [CrossRef] [PubMed]
  3. McManus, D.P.; Dunne, D.W.; Sacko, M.; Utzinger, J.; Vennervald, B.J.; Zhou, X.-N. Schistosomiasis. Nat. Rev. Dis. Primers 2018, 4, 13–31. [Google Scholar] [CrossRef] [PubMed]
  4. World Health Organization (WHO). Ending the Neglect to Attain the Sustainable Development Goals: A Road Map for Neglected Tropical Diseases 2021–2030; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  5. Wang, W.; Wang, L.; Liang, Y.S. Susceptibility or resistance of praziquantel in human schistosomiasis: A review. Parasitol. Res. 2012, 111, 1871–1877. [Google Scholar] [CrossRef] [PubMed]
  6. Caldwell, N.; Afshar, R.; Baragaña, B.; Bustinduy, A.L.; Caffrey, C.R.; Collins, J.J.; Fusco, D.; Garba, A.; Gardner, M.; Gomes, M.; et al. Perspective on Schistosomiasis Drug Discovery: Highlights from a Schistosomiasis Drug Discovery Workshop at Wellcome Collection, London, September 2022. ACS Infect. Dis. 2023, 9, 1046–1055. [Google Scholar] [CrossRef]
  7. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed]
  8. Malheiros, A.; Filho, V.D.; Schmitt, C.B.; Yunes, R.A.; Escalante, A.; Svetaz, L.; Zacchino, S.; Monache, F.R. Antifungal Activity of Drimane Sesquiterpenes from Drimys brasiliensis Using Bioassay-guided Fractionation. J. Pharm. Pharm. Sci. 2005, 8, 335–339. [Google Scholar] [PubMed]
  9. Claudino, V.; Silva, K.; Filho, V.; Yunes, R.; Monache, F.D.; Giménez, A.; Salamanca, E.; Yapu-Gutierrez, D.; Malheiros, A. Drimanes from Drimys brasiliensis with Leishmanicidal and Antimalarial Activity. Mem. Inst. Oswaldo Cruz 2013, 108, 140–144. [Google Scholar] [CrossRef]
  10. Ferreira, B.A.; Filho, A.F.N.; Deconte, S.R.; Tomiosso, T.C.; Thevenard, F.; Andrade, S.P.; Lago, J.H.G.; Araújo, F.D.A. Sesquiterpene Polygodial from Drimys brasiliensis (Winteraceae) Down-Regulates Implant-Induced Inflammation and Fibrogenesis in Mice. J. Nat. Prod. 2020, 83, 3698–3705. [Google Scholar] [CrossRef]
  11. Filho, V.C.; Schlemper, V.; Santos, A.R.S.; Pinheiro, T.R.; Yunes, R.A.; Mendes, G.L.; Calixto, G.L.M.; Monache, F.D. Isolation and Identification of Active Compounds from Drimys winteri Barks. J. Ethnopharmacol. 1998, 62, 223–227. [Google Scholar] [CrossRef] [PubMed]
  12. Freitas, J.; De Lírio, E.J.; Alves-Araújo, A. Flora of Espírito Santo: Winteraceae. Rodriguesia 2022, 73, e200372021. [Google Scholar] [CrossRef]
  13. Corrêa, D.S.; Tempone, A.G.; Reimão, J.Q.; Taniwaki, N.N.; Romoff, P.; Fávero, O.A.; Sartorelli, P.; Mecchi, M.C.; Lago, J.H.G. Anti-leishmanial and Anti-trypanosomal Potential of Polygodial Isolated from Stem Barks of Drimys brasiliensis Miers (Winteraceae). Parasitol. Res. 2011, 109, 231–236. [Google Scholar] [CrossRef]
  14. Simonsen, H.T.; Adsersen, A.; Bremner, P.; Heinrich, M.; Smitt, U.W.; Jaroszewski, J.W. Antifungal Constituents of Melicope borbonica. Phytother. Res. 2004, 18, 542–545. [Google Scholar] [CrossRef] [PubMed]
  15. Mugumbate, G.; Overington, J.P. The Relationship Between Target-class and the Physicochemical Properties of Anti-bacterial Drugs. Bioorg. Med. Chem. 2015, 23, 5218–5224. [Google Scholar] [CrossRef]
  16. Lago, E.M.; Xavier, R.P.; Teixeira, T.R.; Silva, L.M.; da Silva Filho, A.A.; de Moraes, J. Antischistosomal agents: State of art and perspectives. Future Med. Chem. 2018, 10, 89–120. [Google Scholar] [CrossRef] [PubMed]
  17. Sarkar, D.; De Sarkar, S.; Gille, L.; Chatterjee, M. Ascaridole Exerts the Leishmanicidal Activity by Inhibiting Parasite Glycolysis: Anti-leishmanial Activity of Ascaridole. Phytomedicine 2022, 103, 154221. [Google Scholar] [CrossRef] [PubMed]
  18. Brito, J.R.; Wilairatana, P.; Roquini, D.B.; Parra, B.C.; Gonçalves, M.M.; Souza, D.C.S.; Ferreira, E.A.; Salvadori, M.C.; Teixeira, F.S.; Lago, J.H.G.; et al. Neolignans Isolated from Saururus cernuus L. (Saururaceae) Exhibit Efficacy Against Schistosoma mansoni. Sci. Rep. 2022, 12, 19320. [Google Scholar] [CrossRef]
  19. Mona, M.H.; Omran, N.E.; Mansoor, M.A.; El-Fakharany, Z.M. Antischistosomal Effect of Holothurin Extracted from Some Egyptian Sea Cucumbers. Pharm. Biol. 2012, 50, 1144–1150. [Google Scholar] [CrossRef]
  20. Kanyanta, M.; Lengwe, C.; Mambwe, D.; Francisco, K.R.; Liu, L.J.; Uli Sun, Y.; Amarasinghe, D.K.; Caffrey, C.R.; Mubanga Cheuka, P. Activity of N-phenylbenzamide Analogs Against the Neglected Disease Pathogen, Schistosoma mansoni. Bioorg. Med. Chem. Lett. 2023, 82, 129164. [Google Scholar] [CrossRef]
  21. Rocha, B.; de Morais, L.A.; Viana, M.C.; Carneiro, G. Promising Strategies for Improving Oral Bioavailability of Poor Water-Soluble Drugs. Expert Opin. Drug Discov. 2023, 18, 615–627. [Google Scholar] [CrossRef]
  22. Mengarda, A.C.; Silva, M.P.; Cirino, M.E.; Morais, T.R.; Conserva, G.A.A.; Lago, J.H.G.; de Moraes, J. Licarin A, a Neolignan Isolated from Nectandra oppositifolia Nees & Mart. (Lauraceae), Exhibited Moderate Preclinical Efficacy Against Schistosoma mansoni Infection. Phytoter. Res. 2021, 35, 4627–5329. [Google Scholar]
  23. Silva, M.P.; Oliveira, R.M.; Mengarda, A.C.; Roquini, D.B.; Allegretti, S.M.; Salvadori, M.C.; Teixeira, F.S.; de Sousa, D.P.; Pinto, P.L.S.; da Silva Filho, A.A.; et al. Antiparasitic Activity of Nerolidol in a Mouse Model of Schistosomiasis. Int. J. Antimicrob. Agents 2017, 50, 467–472. [Google Scholar] [CrossRef]
  24. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [PubMed]
  25. Costa, D.S.; Leal, C.M.; Cajas, R.A.; Gazolla, M.C.; Silva, L.M.; Carvalho, L.S.A.; Lemes, B.L.; de Moura, R.O.; de Almeida, J.; de Moraes, J.; et al. Antiparasitic Properties of 4-nerolidylcatechol from Pothomorphe umbellata (L.) Miq. (Piperaceae) in vitro and in mice Models with Either Prepatent or Patent Schistosoma mansoni Infections. J. Ethnopharmacol. 2023, 313, 116607. [Google Scholar] [CrossRef] [PubMed]
  26. Hunt, P.R. The C. elegans Model in Toxicity Testing. J. Appl. Toxicol. 2017, 37, 50–59. [Google Scholar] [CrossRef] [PubMed]
  27. Ryan, K.T.; Wheeler, N.J.; Kamara, I.K.; Johnson, H.; Humphries, J.E.; Zamanian, M.; Chan, J.D. Phenotypic Profiling of Macrocyclic Lactones on Parasitic Schistosoma Flatworms. Antimicrob. Agents Chemother. 2023, 67, e0123022. [Google Scholar] [CrossRef] [PubMed]
  28. Jatsa, H.B.; Ngo-Sock, E.T.; Tchuente, L.A.T.; Kamtchouing, P. Evaluation of the In Vivo Activity of Different Concentrations of Clerodendrum umbellatum Poir Against Schistosoma mansoni Infection in Mice. Afr. J. Tradit. Complement. Altern. Med. 2009, 6, 216–221. [Google Scholar] [CrossRef]
  29. Kittur, N.; Castleman, J.D.; Campbell, C.H.; King, C.H.; Colley, D.G. Comparison of Schistosoma mansoni Prevalence and Intensity of Infection, as Determined by the Circulating Cathodic Antigen Urine Assay or by the Kato-Katz Fecal Assay: A Systematic Review. Am. J. Trop. Med. Hyg. 2016, 94, 605–610. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of EDBD (3,6-epidioxy-bisabola-1,10-diene), isolated from the hexane extract of branches of D. brasiliensis.
Figure 1. Chemical structure of EDBD (3,6-epidioxy-bisabola-1,10-diene), isolated from the hexane extract of branches of D. brasiliensis.
Antibiotics 13 00779 g001
Figure 2. Effect of EDBD (3,6-epidioxy-bisabola-1,10-diene) on the viability of S. mansoni and Vero cells. Adult schistosomes and cells were incubated for 72 h in the presence of different concentrations of EDBD. RPMI 1640 medium with 0.5% DMSO, representing the highest concentration of solvent, was used as the negative control. Values are mean (±SD) from three independent experiments performed in triplicate.
Figure 2. Effect of EDBD (3,6-epidioxy-bisabola-1,10-diene) on the viability of S. mansoni and Vero cells. Adult schistosomes and cells were incubated for 72 h in the presence of different concentrations of EDBD. RPMI 1640 medium with 0.5% DMSO, representing the highest concentration of solvent, was used as the negative control. Values are mean (±SD) from three independent experiments performed in triplicate.
Antibiotics 13 00779 g002
Figure 3. Bioavailability radar of EDBD (3,6-epidioxy-bisabola-1,10-diene—left) and PZQ (praziquantel—right).
Figure 3. Bioavailability radar of EDBD (3,6-epidioxy-bisabola-1,10-diene—left) and PZQ (praziquantel—right).
Antibiotics 13 00779 g003
Figure 4. Effect of 3,6-epidioxy-bisabola-1,10-diene (EDBD) and praziquantel (PZQ) on worm burden in S. mansoni-infected mice. Mice were infected with S. mansoni, and forty-two days later, they received a single oral gavage dose of either the test compounds (400 mg/kg) or the vehicle (control). Fifty-six days post-infection, all animals were euthanized, dissected, and all schistosomes were removed, sexed, and counted. Points represent data from individual animals (n = 5 per group). Data are presented as the mean ± SD from five animals per group (n = 5). ** p < 0.01; *** p < 0.001 compared with the infected vehicle-treated control.
Figure 4. Effect of 3,6-epidioxy-bisabola-1,10-diene (EDBD) and praziquantel (PZQ) on worm burden in S. mansoni-infected mice. Mice were infected with S. mansoni, and forty-two days later, they received a single oral gavage dose of either the test compounds (400 mg/kg) or the vehicle (control). Fifty-six days post-infection, all animals were euthanized, dissected, and all schistosomes were removed, sexed, and counted. Points represent data from individual animals (n = 5 per group). Data are presented as the mean ± SD from five animals per group (n = 5). ** p < 0.01; *** p < 0.001 compared with the infected vehicle-treated control.
Antibiotics 13 00779 g004
Figure 5. Effect of 3,6-epidioxy-bisabola-1,10-diene (EDBD) and praziquantel (PZQ) on egg burden in S. mansoni-infected mice. Forty-two days after infecting mice with S. mansoni, a single oral gavage dose (400 mg/kg) of either the test compounds or the vehicle (control) was administered. Mice were euthanized and dissected 56 days post-infection. Egg burden was assessed in both feces and intestinal tissue, with the tissue burden based on the number of immature eggs. Data points represent individual animals (n = 5 per group). The results are presented as the mean ± SD for each group (n = 5). Compared to the infected vehicle-treated control group, both compounds significantly reduced the egg burden (*** p < 0.001).
Figure 5. Effect of 3,6-epidioxy-bisabola-1,10-diene (EDBD) and praziquantel (PZQ) on egg burden in S. mansoni-infected mice. Forty-two days after infecting mice with S. mansoni, a single oral gavage dose (400 mg/kg) of either the test compounds or the vehicle (control) was administered. Mice were euthanized and dissected 56 days post-infection. Egg burden was assessed in both feces and intestinal tissue, with the tissue burden based on the number of immature eggs. Data points represent individual animals (n = 5 per group). The results are presented as the mean ± SD for each group (n = 5). Compared to the infected vehicle-treated control group, both compounds significantly reduced the egg burden (*** p < 0.001).
Antibiotics 13 00779 g005
Table 1. In vitro antischistosomal activities and toxicity of EDBD (3,6-epidioxy-bisabola-1,10-diene) and positive control PZQ (praziquantel).
Table 1. In vitro antischistosomal activities and toxicity of EDBD (3,6-epidioxy-bisabola-1,10-diene) and positive control PZQ (praziquantel).
CompoundS. mansoni Male
EC50/μM
S. mansoni Female
EC50/μM
Vero Cell
CC50/μM
SI a
(Male Worms)
SI a
(Female Worms)
EDBD4.1 ± 1.24.1 ± 1.2˃200˃48˃48
PZQ1.1 ± 0.71.3 ± 0.8˃200˃181˃153
a Selectivity index (SI) = CC50/EC50. Values are means (±SD) of three independent experiments performed in triplicate.
Table 2. In silico physicochemical and ADME parameters of EDBD (3,6-epidioxy-bisabola-1,10-diene) and positive control PZQ (praziquantel).
Table 2. In silico physicochemical and ADME parameters of EDBD (3,6-epidioxy-bisabola-1,10-diene) and positive control PZQ (praziquantel).
ParametersEDBDPZQ
MW236.35 Da312.41 Da
TPSA18.46 Å40.62 Å
Log Po/w3.793.00
Log S−3.21−3.52
Gastroinstestinal absorptionHighHigh
BBBYesYes
Inhibitor CYP1A2NoNo
Inhibitor CYP2C19YesYes
Inhibitor CYP2C9YesNo
Inhibitor CYP2D6NoYes
Inhibitor CYP3A4NoYes
LipinksiYesYes
GhoseYesYes
VeberYesYes
EganYesYes
MueggeYesYes
Bioavaliability Score0.550.55
LeadlikenessYesYes
PAINS0 alert0 alert
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Umehara, E.; Teixeira, T.R.; Cajás, R.A.; Amaro, M.C.; de Moraes, J.; Lago, J.H.G. EDBD—3,6-Epidioxy-1,10-Bisaboladiene—An Endoperoxide Sesquiterpene Obtained from Drimys brasiliensis (Winteraceae) Exhibited Potent Preclinical Efficacy against Schistosoma mansoni Infection. Antibiotics 2024, 13, 779. https://doi.org/10.3390/antibiotics13080779

AMA Style

Umehara E, Teixeira TR, Cajás RA, Amaro MC, de Moraes J, Lago JHG. EDBD—3,6-Epidioxy-1,10-Bisaboladiene—An Endoperoxide Sesquiterpene Obtained from Drimys brasiliensis (Winteraceae) Exhibited Potent Preclinical Efficacy against Schistosoma mansoni Infection. Antibiotics. 2024; 13(8):779. https://doi.org/10.3390/antibiotics13080779

Chicago/Turabian Style

Umehara, Eric, Thainá R. Teixeira, Rayssa A. Cajás, Monique C. Amaro, Josué de Moraes, and João Henrique G. Lago. 2024. "EDBD—3,6-Epidioxy-1,10-Bisaboladiene—An Endoperoxide Sesquiterpene Obtained from Drimys brasiliensis (Winteraceae) Exhibited Potent Preclinical Efficacy against Schistosoma mansoni Infection" Antibiotics 13, no. 8: 779. https://doi.org/10.3390/antibiotics13080779

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