Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Apr 4.
Published in final edited form as: Bioorg Med Chem Lett. 2023 Jan 3;81:129123. doi: 10.1016/j.bmcl.2023.129123

Structure-Activity Relationship of Dibenzylideneacetone Analogs Against the Neglected Disease Pathogen, Trypanosoma brucei

Karol R Francisco a,*, Ludovica Monti a, Wenqian Yang a, Hayoung Park a, Lawrence J Liu a, Kaitlyn Watkins a, Dilini K Amarasinghe a, Marianna Nalli b, Carlos Roberto Polaquini c, Luis O Regasini c, Antônio Eduardo Miller Crotti d, Romano Silvestri b, Lizandra Guidi Magalhães e, Conor R Caffrey a,*
PMCID: PMC10072319  NIHMSID: NIHMS1879274  PMID: 36608774

Abstract

Trypanosoma brucei is a protozoan parasite that causes Human African Trypanosomiasis (HAT), a neglected tropical disease (NTD) that is endemic in 36 countries in sub-Saharan Africa. Only a handful drugs are available for treatment, and these have limitations, including toxicity and drug resistance. Using the natural product, curcumin, as a starting point, several curcuminoids and related analogs were evaluated against bloodstream forms of T. b. brucei. A particular subset of dibenzylideneacetone (DBA) compounds exhibited potent in vitro antitrypanosomal activity with sub-micromolar EC50 values. A structure-activity relationship study including 26 DBA analogs was initiated, and several compounds exhibited EC50 values as low as 200 nM. Cytotoxicity counter screens in HEK293 cells identified several compounds having selectivity indices above 10. These data suggest that DBAs offer starting points for a new small molecule therapy of HAT.

Keywords: Trypanosoma brucei, antiparasitic activity, cytotoxicity, curcuminoid, dibenzylideneacetone

Graphical Abstract

graphic file with name nihms-1879274-f0001.jpg


Human African Trypanosomiasis (HAT), also known as sleeping sickness, is a neglected tropical disease (NTD) caused by the protozoan parasite Trypanosoma brucei.1 There are three subspecies of T. brucei, which are all transmitted by the bite of an infected tsetse fly: T. b. brucei which primarily infects cattle and other animals, and T. b. gambiense and T. b. rhodesiense, which cause HAT.1, 2 HAT is endemic in sub-Saharan Africa with approximately 55 million people at risk of infection. 2, 3 The disease occurs in two stages: the initial, and hemolymphatic, stage 1, and the stage 2 infection that affects the central nervous system (CNS).2 Current drug treatments for HAT include pentamidine and suramin, which are used as treatments for stage 1 infection by T. b. gambiense and T. b. rhodesiense, respectively.4 For stage 2 infections, the drugs traditionally used, e.g., melarsoprol, and eflornithine combined with nifurtimox, can induce severe adverse events, must be administered intravenously under medical supervision, and are associated with drug resistance.411 Recently, fexinidazole, which treats both stage 1 and 2 infection by T. b. gambiense, was approved as the first oral drug against HAT.12, 13 Nevertheless, with the parasite’s ability to evolve resistance,11, 14 new therapies should be identified.

Curcumin (1) is a natural product isolated from the perennial herb, Curcuma longa L. (turmeric), and is a traditional ethnomedicine.15, 16 First discovered in the early 19th century, curcumin and structurally related curcuminoids made their way to modern medicine in the mid- to late-20th century,15 and are biologically active in a variety of biomedical applications,17, 18 including as anticancer,1922 anti-inflammatory,23, 24 and antiparasitic agents.2528 In spite of its utility, curcumin has been described as a pan-assay interference (PAIN)29 and a poor lead compound30, 31 due to its instability,32, 33 as well as its poor adsorption, distribution, metabolism, excretion and toxicological/pharmacokinetic (ADMET/PK) properties.3336

Structurally, curcumin is a symmetrical molecule characterized by aryl rings separated by an α,β-unsaturated β-diketone linker. In vivo, curcuminoids are reduced to hexahydrocurcuminoids and form conjugates with sulfate and glucuronic acid.32, 37, 38 In vitro, one of the key degradation pathways of curcuminoids occurs via autoxidation of the α,β-unsaturated β-diketone linker to a bicyclopentadione 2 (Figure 1A).32, 39 To improve the in vitro and in vivo stability of curcumin and related curcuminoids, studies have explored derivatives in which the site of curcuminoid degradation has been modified.40 Structurally derived from the curcuminoid backbone, dibenzylideneacetone (DBA, 3, Figure 1B) compounds bear α,β-unsaturated ketone aryl linkers that provide for improved in vitro stability and in vivo PK relative to curcumin.40 Interestingly, DBA compounds are structurally similar to chalcones (e.g., 4, Figure 1B), which are considered privileged structures in medicinal chemistry and are a component of some marketed drugs.41, 42 DBA analogs have antiparasitic activity, including against Leishmania donovani,43, 44 Leishmania amazonensis,45 Plasmodium falciparum,4648 Trichomonas vaginalis49 and Trypanosoma cruzi.45, 5052 Despite the continued interest in DBAs, their antiparasitic activity against Trypanosoma brucei, including any associated structure-activity relationship (SAR), has not been reported.

Figure 1.

Figure 1

(A) Chemical structure of curcumin and its major degradation product. (B) Chemical structures of DBA and chalcone.

Here, several natural and synthetic curcuminoids5358 were evaluated in vitro against bloodstream forms of T. b. brucei Lister 427. Also, curcuminoid analogs, including 26 DBAs, which bear alternative aryl linkers, were explored and evaluated. Finally, all compounds were evaluated for cytotoxicity against human embryonic kidney (HEK)293 cells. Test compounds (3, 547) were synthesized as described previously.5358 Curcumin (1) and the positive control, pentamidine, were purchased from Sigma-Aldrich. The purity of all compounds was ≥95% as determined by LC/MS.

All compounds (1, 3, 547) were first tested at a concentration of 4 μM against bloodstream forms of T. b. brucei Lister 427, and at a concentration of 20 μM for cytotoxicity against HEK293 cells, using the SYBR green59, 60 and resazurin61 cell viability assays, respectively (see Supporting Information for assay details and conditions). Compounds that were sufficiently active at these concentrations, i.e., > 50% growth inhibition, were then tested in an eight-point dose-response assay. The results are summarized in Tables 13.

Table 1.

Activity of curcumin and curcuminoids against T. b. brucei and HEK293 cells.

graphic file with name nihms-1879274-t0006.jpg
Cpd ID Structure R T. b. brucei EC50 (μM)a HEK293 CC50 (μM)b Cpd ID Structure R T. b. brucei EC50 (μM) a HEK293 CC50 (μM)b
1 graphic file with name nihms-1879274-t0007.jpg 1.78 ± 0.03 5.56 ± 0.05 9 graphic file with name nihms-1879274-t0008.jpg >4 >20
5 graphic file with name nihms-1879274-t0009.jpg >4 >20 10 graphic file with name nihms-1879274-t0010.jpg 3.99 ± 1.56 5.56 ± 0.05
6 graphic file with name nihms-1879274-t0011.jpg 3.29 ± 0.12 6.73 ± 0.77 11 graphic file with name nihms-1879274-t0012.jpg >4 >20
7 graphic file with name nihms-1879274-t0013.jpg >4 >20 12 graphic file with name nihms-1879274-t0014.jpg >4 >20
8 graphic file with name nihms-1879274-t0015.jpg >4 >20 Pentamidine c graphic file with name nihms-1879274-t0016.jpg 0.012 ± 0.002 >4
a

Antitrypanosomal activity against T. b. brucei Lister 427 parasites was assessed using the SYBR green cell viability assay after 72 h.

b

Cytotoxicity of test compounds against HEK293 cells was assessed using the resazurin cell viability assay after 48 h. At least three assay replicates were performed each in duplicate. Means ± SD values are shown.

c

Pentamidine was used as the positive drug control.

Table 3.

Activity of symmetrical and asymmetrical DBA analogs against T. b. brucei and HEK293 cells.

graphic file with name nihms-1879274-t0038.jpg
Cpd ID Structure T. b. brucei EC50 (μM)a HEK293 CC50 (μM)b
R R’
3 graphic file with name nihms-1879274-t0039.jpg graphic file with name nihms-1879274-t0040.jpg 0.80 ± 0.20 8.33 ± 0.48
28 graphic file with name nihms-1879274-t0041.jpg graphic file with name nihms-1879274-t0042.jpg >4 >20
29 graphic file with name nihms-1879274-t0043.jpg graphic file with name nihms-1879274-t0044.jpg >4 >20
30 graphic file with name nihms-1879274-t0045.jpg graphic file with name nihms-1879274-t0046.jpg 0.24 ± 0.07 1.45 ± 0.11
31 graphic file with name nihms-1879274-t0047.jpg graphic file with name nihms-1879274-t0048.jpg 0.84 ± 0.20 9.74 ± 1.20
32 graphic file with name nihms-1879274-t0049.jpg graphic file with name nihms-1879274-t0050.jpg 1.42 ± 0.35 7.64 ± 1.90
33 graphic file with name nihms-1879274-t0051.jpg 0.20 ± 0.06 3.58 ± 0.60
34 graphic file with name nihms-1879274-t0052.jpg >4 3.70 ± 0.08
35 graphic file with name nihms-1879274-t0053.jpg graphic file with name nihms-1879274-t0054.jpg >4 5.49 ± 0.22
36 graphic file with name nihms-1879274-t0055.jpg 0.29 ± 0.03 5.79 ± 0.35
37 graphic file with name nihms-1879274-t0056.jpg 1.25 ± 0.09 5.85 ± 0.60
38 graphic file with name nihms-1879274-t0057.jpg 0.37 ± 0.06 5.54 ± 0.56
39 graphic file with name nihms-1879274-t0058.jpg 0.66 ± 0.15 5.63 ± 0.36
40 graphic file with name nihms-1879274-t0059.jpg >4 9.79 ± 0.22
41 graphic file with name nihms-1879274-t0060.jpg 3.74 ± 0.30 3.70 ± 0.10
42 graphic file with name nihms-1879274-t0061.jpg 1.93 ± 0.48 >20
43 graphic file with name nihms-1879274-t0062.jpg 2.15 ± 0.12 11.35 ± 0.48
44 graphic file with name nihms-1879274-t0063.jpg 2.33 ± 0.38 7.05 ± 0.92
45 graphic file with name nihms-1879274-t0064.jpg 2.51 ± 0.39 >20
46 graphic file with name nihms-1879274-t0065.jpg 0.20 ± 0.03 2.88 ± 0.86
47 graphic file with name nihms-1879274-t0066.jpg graphic file with name nihms-1879274-t0067.jpg 0.41 ± 0.02 6.61 ± 0.90
Pentamidine c graphic file with name nihms-1879274-t0068.jpg 0.012 ± 0.002 >4
a-c

See Table 1 footnote.

As previously reported,62, 63 curcumin exhibited low micromolar activity against bloodstream forms of T. b. brucei with a 50% effective concentration (EC50) of 1.78 μM (Table 1; published EC50 values for curcumin are 2.5 μM62 and 2.7 μM63). Modifications around the aryl rings led to curcuminoids 512, that were active (EC50 values <4 μM) or inactive (EC50 >4 μM) against the parasite. Removal of 3-methoxy and 4-hydroxy from 1 to 5 resulted in no measurable activity in first pass screens. Likewise, aryl substitutions with 3-methoxy (7), 3,4-dimethoxy (8) and 2,4-dimethoxy (9) were inactive. Interestingly, derivatives with more electron rich aryl rings, i.e., 6 and 10¸ exhibited activity against T. b. brucei with EC50 values of 3.29 and 3.99 μM, respectively. Curcuminoids bearing one or more electron withdrawing groups on the aryl rings (11 and 12) were inactive. Overall, the SAR suggests a preference for curcuminoids with highly electron-rich aryl rings; however, as these compounds exhibited only moderate or no activity against T. b. brucei compared to the positive control drug, pentamidine (EC50 = 0.012 μM), they were not pursued further.

Next, a focused set of 15 analogs with alternative aryl linkers were explored, including those derived from dibenzylideneacetone (DBA), and its reduced derivatives, 1,5-diphenylpentan-3-one (DPN) and 1,5-diphenylpentan-3-ol (DPL). These included five sets of matched molecular pairs that bear the same aryl substitutions and vary only by the oxidation state of the linker (Table 2). From this set, three compounds, 13, 16 and 19, displayed sub-micromolar activity against T. b. brucei. All three compounds belonged to the DBA class, whereas their corresponding DPN and DPL analogs were inactive at 4 μM. Compounds were also tested for cytotoxicity against mammalian HEK293 cells. Although 13, 16 and 19 showed some cytotoxicity with CC50 values between 0.30 and 4.55 μM, 16 and 19 were more active against T. b. brucei than HEK293 cells with selectivity indices (SI = HEK293 CC50/T. b. brucei EC50) above 10.

Table 2.

Activity of dibenzylideneacetone, 1,5-diphenylpentan-3-one and 1,5-diphenylpentan-3-ol analogs against T. b. brucei and HEK293 cells.

Cpd ID Structure T. b. brucei EC50 (μM)a HEK293 CC50 (μM)b
Linker R
13 graphic file with name nihms-1879274-t0017.jpg graphic file with name nihms-1879274-t0018.jpg 0.36 ± 0.06 0.30 ± 0.03
14 graphic file with name nihms-1879274-t0019.jpg >4 >20
15 graphic file with name nihms-1879274-t0020.jpg >4 >20
16 graphic file with name nihms-1879274-t0021.jpg graphic file with name nihms-1879274-t0022.jpg 0.42 ± 0.02 4.55 ± 0.17
17 graphic file with name nihms-1879274-t0023.jpg >4 >20
18 graphic file with name nihms-1879274-t0024.jpg >4 >20
19 graphic file with name nihms-1879274-t0025.jpg graphic file with name nihms-1879274-t0026.jpg 0.31 ± 0.04 4.27 ± 0.18
20 graphic file with name nihms-1879274-t0027.jpg >4 >20
21 graphic file with name nihms-1879274-t0028.jpg >4 >20
22 graphic file with name nihms-1879274-t0029.jpg graphic file with name nihms-1879274-t0030.jpg >4 >20
23 graphic file with name nihms-1879274-t0031.jpg >4 >20
24 graphic file with name nihms-1879274-t0032.jpg >4 >20
25 graphic file with name nihms-1879274-t0033.jpg graphic file with name nihms-1879274-t0034.jpg >4 >20
26 graphic file with name nihms-1879274-t0035.jpg >4 >20
27 graphic file with name nihms-1879274-t0036.jpg >4 >20
Pentamidine c graphic file with name nihms-1879274-t0037.jpg 0.012 ± 0.002 >4
a-c

See Table 1 footnote.

Additional DBA analogs were explored and tested against T. b. brucei, including compounds that are either structurally symmetrical (3, 2834) or asymmetrical (3547) (Table 3). DBA 3, which bears symmetrical, unsubstituted aryl rings, was active with an EC50 value of 0.80 μM. In contrast, the symmetrical derivatives 3-methoxy (28) and 4-methoxy (29) compounds were inactive at 4 μM. Interestingly, whereas 30 (3,3-dimethoxy) exhibited good activity with an EC50 value of 0.24 μM, its analog 34, which has similar substitutions but bears a cyclohexane ring at the DBA linker, was inactive.

Regarding the asymmetrical DBA analogs (3547), 11 of the 13 compounds exhibited moderate (EC50 values between 1 and 4 μM) to potent (EC50 <1 μM) antitrypanosomal activity. In general, asymmetrical compounds with electron rich rings (3639) exhibited good activity against T. b. brucei with EC50 values ranging from 0.29 to 1.25 μM. Exceptions to this pattern included compounds 35 and 40, which, despite having electron-rich aryl rings, showed no activity. Conversely, asymmetrical analogs with electron deficient rings (4245) showed comparatively less activity with EC50 values ranging from 1.93 to 2.51 μM. Interestingly, the asymmetrical DBA, 46, which bears an electron-withdrawing nitro group on one aryl ring, was one of the most potent analogs with an EC50 value of 0.20 μM. Overall, the SAR of the DBA analogs suggests that the electronic properties of the aryl rings may influence potency against T. b. brucei, and that there may also be some preference for certain functional groups, particularly a nitro (46), that is not necessarily dictated by the electronic properties of the aryl rings. All DBA analogs were tested for cytotoxicity against HEK293 cells. In general, the 19 antitrypanosomal DBA analogs exhibited some cytotoxicity, though the most active compounds were more active against T. b. brucei with 17 analogs having SI values above 3, and 10 analogs with SI values above 10 (Figure 2).

Figure 2.

Figure 2.

Plot of the log of T. b. brucei EC50 values vs. HEK293 CC50 values. Indicated are those DBA analogs that exhibit SI values above 1 (red line), 3 (blue line) and 10 (black line).

To measure the speed of the antitrypanosomal activity of the DBA analogs, a time-kill experiment was conducted using 46. T. b. brucei parasites were incubated with 46 (EC99 = 0.90 ± 0.16 μM) at 1, 2 and 4 μM. As a comparison, parasites were also incubated with the positive control drug, pentamidine, at 0.5, 1 and 2 μM (Figure 3). Compound 46 required less than 24 h (less than 6 h at 2 and 4 μM) to kill the parasite at all concentrations tested, whereas pentamidine required >30 h to kill all of the parasites (Figure 3).

Figure 3.

Figure 3.

Compound 46 is a faster-acting antitrypanosomal compound than pentamidine. Parasites were quantified by hemocytometer cell counting and the counts were normalized to a negative (DMSO) control. Means of three replicates ± SD bars are shown.

To distinguish trypanocidal activity from trypanostatic activity, we performed a washout experiment using compound 46. We first incubated 1, 2 and 4 μM of the compound with trypanosomes for 6, 4 and 2 h, respectively, and then washed the remaining parasites three times. The parasites did not regain the ability to divide after monitoring for a further 48 h, suggesting that 46 is trypanocidal rather than trypanostatic (Figure S1, Supporting Information).

Curcumin and curcuminoid analogs have established biological activity, including as anticancer,1922 anti-inflammatory,23, 24 and antiparasitic agents.2528 Nonetheless, these natural products have been considered to be poor lead compounds2931 due to their instability32, 33 and weak ADMET/PK properties.3336 In the present study, curcumin (1) and curcuminoids (512; Table 1) were evaluated for their antiparasitic activity against bloodstream forms of T. b. brucei. Whereas curcumin 1, and curcuminoids 6 and 10, resulted in moderate antitrypanosomal activity (EC50 values of 1–4 μM), the remaining eight curcuminoids were inactive (EC50 values >4 μM). To explore structurally related derivatives with improved metabolic stability,40 compounds with alternative aryl linkers were explored, including DBAs and their reduced analogs, DPN and DPL (Table 2). Through the matched molecular pair analysis of the DBA, DPN and DPL analogs, it was established that the presence of the dienone in DBA, which may serve as a Michael acceptor, is essential for antitrypanosomal activity. This is demonstrated by the loss of activity when the Michael acceptor is removed, e.g., 13 with an EC50 value of 0.36 μM vs 14 and 15 with EC50 values >4 μM (Table 2).

Compounds with Michael acceptors are known antiparasitics.50, 6264 Due to the intrinsic reactivity of Michael acceptors, a possible MOA may the involve reaction with nucleophilic residues, including with the catalytic site of cysteine proteases. A study by Ettari et al. has shown that curcumin, in combination with related Curcuma longa L. natural product, genistein, inhibits the major cathepsin L-like cysteine protease (TbrCATL)65 of T. b. rhodesiense.66 In the context of mono-enones, studies by Changtam et al. explored curcuminoid analogs bearing Michael acceptor mono-enones, several of which were active against bloodstream forms of T. b. brucei, Leishmania major promastigotes and Leishmania mexicana amastigotes.62 In a follow up study by Alkhaldi et al., the mechanism of action (MOA) of these mono-enones in T. b. brucei was explored, and their antitrypanosomal activity was attributed to the formation of trypanothione adducts as a result of Michael addition between the mono-enone and trypanothione. These mono-enone Michael acceptors may, therefore, deplete the parasite of essential thiols, leading to parasite death.63 For the DBA analogs evaluated here, a similar MOA may be possible (Figure 4).

Figure 4.

Figure 4.

Proposed MOA of Michael acceptors as antitrypanosomal agents,63 the influence of DBA aryl substitutions and reactivity towards Michael addition, and the stability of the Michael adduct that may form.67 TSH = trypanothione; EWG = electron withdrawing group; EDG = electron donating group.

From the present evaluation of 26 Michael acceptor DBA analogs against T. b. brucei, several aspects of the SAR were observed. In particular, the 26 DBA analogs differed by 1) the electronic properties of the aryl rings, 2) their symmetry, and 3), for a small number of compounds, the cyclic nature of the aryl linker (e.g., compounds 33 and 34). Using the unsubstituted DBA 3 as the reference compound, 13, 16, 22, 2831, 3441 and 47 have electron-donating groups on one or both aryl rings, whereas 19, 25, 32, 33 and 4246 have electron-withdrawing groups on one or both aryl rings. The electronic nature of the aryl rings is expected to influence the reactivity towards Michael addition, as well as the stability of the Michael adduct that may form (Figure 4). In general, electron rich analogs were moderately more active (8 out of 16 compounds exhibited EC50 values <1 μM) than electron deficient analogs (3 out of 9 exhibited EC50 values <1 μM). Studies have shown that adducts formed by Michael acceptors bearing electron-rich aryl rings are expected to be more stable and less reversible than those bearing electron-deficient aryl rings,67 which may explain why electron rich DBA analogs were generally more active. Interestingly, despite this overall trend in the SAR, 46, an asymmetrical nitro DBA analog bearing one electron deficient aryl ring, was one of the most active analogs with an EC50 value of 0.20 μM. It is known that nitro-DBAs increase the production of reactive oxygen species (ROS) and damage the parasite antioxidant system in Trypanosoma cruzi.50 This cidal activity against T. cruzi was attributed to the presence of the nitro group within the DBA analog that may be involved in the production of ROS.68 Due to the potent activity of 46 against T. b. brucei, a similar MOA in T. b. brucei is possible.

In addition, the SAR of both symmetrical (3, 13, 16, 19, 22, 25, 2834) and asymmetrical DBAs (3547) was evaluated. As DBA analogs have two alkenes and therefore two sites for Michael addition, the key difference between the symmetrical and asymmetrical analogs is the possible formation of either one or two Michael adducts, respectively (see Figure 4 for an example of two generic Michael adducts). Generally, the symmetrical DBAs were more active (7 out of 13 exhibited EC50 values <1 μM) than the asymmetrical DBAs (5 out of 13 with EC50 values <1 μM). However, upon examining the symmetrical compound, 31, and its structurally closest asymmetrical analog, 39, the asymmetrical analog was slightly more potent against T. b. brucei (respective EC50 values of 0.84 and 0.66 μM). This suggests that further study of the SAR is required to assess whether symmetry contributes to the antitrypanosomal activity of DBA analogs.

Finally, whereas most of the DBA analogs studied have linear dienone aryl linkers, two analogs with cyclic dienones, the piperidinone, 33, and the cyclohexanone, 34, were also included in the study. Compound 33 exhibited one of the most potent activities against T. b. brucei (EC50 value of 0.20 μM) whereas 34 exhibited no activity (EC50 value > 4 μM). Additional exploration of these cyclic dienones is required to evaluate the SAR of linear vs cyclic DBAs.

All compounds were also tested for toxicity against HEK293 cells and those that were active against T. b. brucei tended to have measurable activity against the mammalian cell line, although with a broad range of SI values (Figure 2). These data might suggest a general MOA irrespective of cell type. As per the MOA suggested by Alkhaldi et al. that involves the interaction with trypanothione in trypanosomes, mammalian cells also have essential thiols, not least, glutathione, that could form glutathione adducts with the DBAs.63

To conclude, the present study identifies a series of DBA analogs as potent antitrypanosomal agents with EC50 values as low as 0.20 μM and SI values up to >10. Additionally, whereas the DBA analogs were comparatively less active than pentamidine, the time-to-kill experiments show that analog 46 is a relatively faster trypanocide. The data encourage the continued investigation of DBAs as the possible basis for a novel chemotherapy of HAT.

Supplementary Material

Supporting info

Acknowledgments

KRF is supported by the CARING T32 Training Grant (T32AI007036). Maintenance of T. b. brucei in culture was, in part, supported by NIH-NIAID R21AI141210 and R21AI171824 to CRC among other principal investigators. JChem for Office was used to draw, display and characterize chemical structures and substructures, version 21.13.0.915. ChemAxon (https://www.chemaxon.com). LGM acknowledges the State of São Paulo Research Foundation-FAPESP (Grant numbers: 2016/24456-1 and 2018/50011-2).

References

  • 1.Human African Trypanosomiasis (Sleeping Sickness): The Road to Elimination Revisited—Achievements and Remaining Challenges. Tropical Medicine and Infectious Diseases: MDPI; 2020. [Google Scholar]
  • 2.Ponte-Sucre A An Overview of Trypanosoma brucei Infections: An Intense Host–Parasite Interaction. Front Microbiol. 2016;7: 2126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Trypanosomiasis, human African (sleeping sickness); 2022. https://www.who.int/news-room/fact-sheets/detail/trypanosomiasis-human-african-(sleeping-sickness).
  • 4.Babokhov P, Sanyaolu A, Oyibo W, Fagbenro-Beyioku A, Iriemenam N. A current analysis of chemotherapy strategies for the treatment of human African trypanosomiasis. Pathogens and Global Health. 2013;107(5): 242–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bouteille B, Buguet A. The detection and treatment of human African trypanosomiasis. Res Rep Trop Med. 2012;3: 35–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fairlamb AH, Horn D. Melarsoprol Resistance in African Trypanosomiasis. Trends Parasitol. 2018;34(6): 481–492. [DOI] [PubMed] [Google Scholar]
  • 7.Baker N, de Koning HP, Mäser P, Horn D. Drug resistance in African trypanosomiasis: the melarsoprol and pentamidine story. Trends Parasitol. 2013;29(3): 110–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Barrett MP, Vincent IM, Burchmore RJ, Kazibwe AJ, Matovu E. Drug resistance in human African trypanosomiasis. Future Microbiol. 2011;6(9): 1037–1047. [DOI] [PubMed] [Google Scholar]
  • 9.Sokolova AY, Wyllie S, Patterson S, Oza SL, Read KD, Fairlamb AH. Cross-resistance to nitro drugs and implications for treatment of human African trypanosomiasis. Antimicrob Agents Chemother. 2010;54(7): 2893–2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Vincent IM, Creek D, Watson DG, Kamleh MA, Woods DJ, Wong PE, Burchmore RJ, Barrett MP. A molecular mechanism for eflornithine resistance in African trypanosomes. PLoS Pathog. 2010;6(11): e1001204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kasozi KI, MacLeod ET, Ntulume I, Welburn SC. An Update on African Trypanocide Pharmaceutics and Resistance. Front Vet Sci. 2022;9: 828111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mesu VKBK Kalonji WM, Bardonneau C Mordt OV, Tete DN, Blesson S, Simon F, Delhomme S, Bernhard S, Mbembo, Moke CM, Vuvu SL, E’kitiak JM, Masa FA, Ilunga MM, Nzambi DMM, Malu TM, Tshilumbwa SK, Bolengi FB, Matsho MN, Lumbala C, Scherrer B, Strub-Wourgaft N, Tarral N. Oral fexinidazole for stage 1 or early stage 2 African Trypanosoma brucei gambiense trypanosomiasis: a prospective, multicentre, open-label, cohort study. Lancet Glob Health. 2021;9(7): e999–e1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dickie EA, Giordani F, Gould MK, Maser P, Burri C, Mottram JC, Rao SPS, Barrett MP. New Drugs for Human African Trypanosomiasis: A Twenty First Century Success Story. Trop Med Infect Dis. 2020;5(1): 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brun R, Don R, Jacobs RT, Wang MZ, Barrett MP. Development of novel drugs for human African trypanosomiasis. Future Microbiol. 2011;6(6): 677–691. [DOI] [PubMed] [Google Scholar]
  • 15.Priyadarsini KI. The chemistry of curcumin: from extraction to therapeutic agent. Molecules. 2014;19(12): 20091–20112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gupta SC, Patchva S, Koh W, Aggarwal BB. Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin Exp Pharmacol Physiol. 2012;39(3): 283–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mishra S, Palanivelu K. The effect of curcumin (turmeric) on Alzheimer’s disease: An overview. Ann Indian Acad Neurol. 2008;11(1): 13–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hewlings SJ, Kalman DS. Curcumin: A Review of Its Effects on Human Health. Foods. 2017;6(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ledda A, Belcaro G, Dugall M, Luzzi R, Scoccianti M, Togni S, Appendino G, Ciammaichella G. Meriva®, a lecithinized curcumin delivery system, in the control of benign prostatic hyperplasia: a pilot, product evaluation registry study. Panminerva Med. 2012;54(1 Suppl 4): 17–22. [PubMed] [Google Scholar]
  • 20.Bayet-Robert M, Kwiatkowski F, Leheurteur M, Gachon F, Planchat E, Abrial C, Mouret-Reynier MA, Durando X, Barthomeuf C, Chollet P. Phase I dose escalation trial of docetaxel plus curcumin in patients with advanced and metastatic breast cancer. Cancer Biol Ther. 2010;9(1): 8–14. [DOI] [PubMed] [Google Scholar]
  • 21.Ghalaut VS, Sangwan L, Dahiya K, Ghalaut PS, Dhankhar R, Saharan R. Effect of imatinib therapy with and without turmeric powder on nitric oxide levels in chronic myeloid leukemia. J Oncol Pharm Pract. 2012;18(2): 186–190. [DOI] [PubMed] [Google Scholar]
  • 22.Tomeh MA, Hadianamrei R, Zhao X. A Review of Curcumin and Its Derivatives as Anticancer Agents. Int J Mol Sci. 2019;20(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sandur SK, Pandey MK, Sung B, Ahn SK, Murakami A, Sethi G, Limtrakul P, Badmaev V, Aggarwal BB. Curcumin, demethoxycurcumin, bisdemethoxycurcumin, tetrahydrocurcumin and turmerones differentially regulate anti-inflammatory and anti-proliferative responses through a ROS-independent mechanism. Carcinogenesis. 2007;28(8): 1765–1773. [DOI] [PubMed] [Google Scholar]
  • 24.Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol. 2009;41(1): 40–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.de Paula Aguiar D, Moscardini MBM, Morais ER, de Paula RG, Ferreira PM, Afonso A, Belo S, Ouchida AT, Curti C, Cunha WR, Rodrigues V, Magalhaes LG. Curcumin Generates Oxidative Stress and Induces Apoptosis in Adult Schistosoma mansoni Worms. PLoS One. 2016;11(11): e0167135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cui L, Miao J. Cytotoxic effect of curcumin on malaria parasite Plasmodium falciparum: inhibition of histone acetylation and generation of reactive oxygen species. Antimicrob Agents Chemother. 2007;51(2): 488–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nagajyothi F, Zhao D, Weiss LM, Tanowitz HB. Curcumin treatment provides protection against Trypanosoma cruzi infection. Parasitol Res. 2012;110(6): 2491–2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gutiérrez-Gutiérrez F, Palomo-Ligas L, Hernández-Hernández JM, Perez-Rangel A, Aguayo-Ortiz R, Hernandez-Campos A, Castillo R, Gonzalez-Pozos S, Cortes-Zarate R, Ramirez-Herrera MA, Mendoza-Magana ML, Castillo-Romero A. Curcumin alters the cytoskeleton and microtubule organization on trophozoites of Giardia lamblia. Acta Trop. 2017;172: 113–121. [DOI] [PubMed] [Google Scholar]
  • 29.Baell J, Walters MA. Chemistry: Chemical con artists foil drug discovery. Nature. 2014;513(7519): 481–483. [DOI] [PubMed] [Google Scholar]
  • 30.Bisson J, McAlpine JB, Friesen JB, Chen SN, Graham J, Pauli GF. Can Invalid Bioactives Undermine Natural Product-Based Drug Discovery? J Med Chem. 2016;59(5): 1671–1690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. The Essential Medicinal Chemistry of Curcumin. J Med Chem. 2017;60(5): 1620–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Schneider C, Gordon ON, Edwards RL, Luis PB. Degradation of Curcumin: From Mechanism to Biological Implications. J Agric Food Chem. 2015;63(35): 7606–7614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Esatbeyoglu T, Ulbrich K, Rehberg C, Rohn S, Rimbach G. Thermal stability, antioxidant, and anti-inflammatory activity of curcumin and its degradation product 4-vinyl guaiacol. Food Funct. 2015;6(3): 887–893. [DOI] [PubMed] [Google Scholar]
  • 34.Ravindranath V, Chandrasekhara N. Absorption and tissue distribution of curcumin in rats. Toxicology. 1980;16(3): 259–265. [DOI] [PubMed] [Google Scholar]
  • 35.Mathews S, Rao M. Interaction of Curcumin with Glutathione. Int J Pharm. 1991;76(3): 257–259. [Google Scholar]
  • 36.Hong D, Son Y, Choi I, Park W. The inhibitory effect of curcumin on voltage-dependent K+ channels in rabbit coronary arterial smooth muscle cells. cells. Biochem Biophys Res Commun. 2013;430(1): 307–312. [DOI] [PubMed] [Google Scholar]
  • 37.Hoehle SI, Pfeiffer E, Sólyom AM, Metzler M. Metabolism of curcuminoids in tissue slices and subcellular fractions from rat liver. J Agric Food Chem. 2006;54(3): 756–764. [DOI] [PubMed] [Google Scholar]
  • 38.Pan MH, Huang TM, Lin JK. Biotransformation of curcumin through reduction and glucuronidation in mice. Drug Metab Dispos. 1999;27(4): 486–494. [PubMed] [Google Scholar]
  • 39.Griesser M, Pistis V, Suzuki T, Tejera N, Pratt DA, Schneider C. Autoxidative and cyclooxygenase-2 catalyzed transformation of the dietary chemopreventive agent curcumin. J Biol Chem. 2011;286(2): 1114–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liang G, Shao L, Wang Y, Zhao C, Chu Y, Xiao J, Zhao Y, Li X, Yang S. Exploration and synthesis of curcumin analogues with improved structural stability both in vitro and in vivo as cytotoxic agents. Bioorg Med Chem. 2009;17(6): 2623–2631. [DOI] [PubMed] [Google Scholar]
  • 41.Zhuang C, Zhang W, Sheng C, Xing C, Miao Z. Chalcone: A Privileged Structure in Medicinal Chemistry. Chem Rev. 2017;117(12): 7762–7810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gomes MN, Muratov EN, Pereira M, Peixoto JC, Rosseto LP, Cravo PVL, Andrade CH, Neves BJ. Chalcone Derivatives: Promising Starting Points for Drug Design. Molecules. 2017;22(8): 1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chauhan IS, Rao GS, Shankar J, Chauhan LKS, Kapadia GJ, Singh N. Chemoprevention of Leishmaniasis: In-vitro antiparasitic activity of dibenzalacetone, a synthetic curcumin analog leads to apoptotic cell death in Leishmania donovani. Parasitol Int. 2018;67(5): 627–636. [DOI] [PubMed] [Google Scholar]
  • 44.Chauhan IS, Marwa S, Rao GS, Singh N. Antiparasitic dibenzalacetone inhibits the GTPase activity of Rab6 protein of Leishmania donovani (LdRab6), a potential target for its antileishmanial effect. Parasitol Res. 2020;119(9): 2991–3003. [DOI] [PubMed] [Google Scholar]
  • 45.Din Z, Fill T, de Assis F, Lazarin-Bidoia D, Kaplum V, Garcia FP, Nakamura CV, de Oliveira KT, Rodrigues-Filho E. Unsymmetrical 1,5-diaryl-3-oxo-1,4-pentadienyls and their evaluation as antiparasitic agents. Bioorg Med Chem. 2014;22(3): 1121–1127. [DOI] [PubMed] [Google Scholar]
  • 46.Aher RB, Wanare G, Kawathekar N, Kumar RR, Kaushik NK, Sahal D, Chauhan VS. Dibenzylideneacetone analogues as novel Plasmodium falciparum inhibitors. Bioorg Med Chem Lett. 2011;21(10): 3034–3036. [DOI] [PubMed] [Google Scholar]
  • 47.Dohutia C, Chetia D, Gogoi K, Sarma K. Design, in silico and in vitro evaluation of curcumin analogues against Plasmodium falciparum. Exp Parasitol. 2017;175: 51–58. [DOI] [PubMed] [Google Scholar]
  • 48.Joshi B, Mohanakrishnan D, Mittal G, Kar S, Pola JK, Golakoti NR, Nanubolu JB, Babu RD, Jumar SSS, Sahal D. Synthesis, mechanistic and synergy studies of diarylidenecyclohexanone derivatives as new antiplasmodial pharmacophores. Med Chem Res. 2018;27(10): 2312–2324. [Google Scholar]
  • 49.Carapina da Silva C, Pacheco BS, das Neves RN, Alves MSD, Sena-Lopes A, Moura S, Borsuk S, de Pereira CMP. Antiparasitic activity of synthetic curcumin monocarbonyl analogues against Trichomonas vaginalis. Biomed Pharmacother. 2019;111: 367–377. [DOI] [PubMed] [Google Scholar]
  • 50.Lazarin-Bidóia D, Desoti VC, Martins SC, Ribeiro FM, Din ZU, Rodrigues-Filho E, Ueda-Nakamura T, Nakamura CV, de Oliveira Silva, S. Dibenzylideneacetones Are Potent Trypanocidal Compounds That Affect the Trypanosoma cruzi Redox System. Antimicrob Agents Chemother. 2016;60(2): 890–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Peron F, Lazarin-Bidoia D, Din ZU, Rodrigues-Filho E, Ueda-Nakamura T, Silva SO, Nakamura CV. Effects of (1E,4E)-2-Methyl-1,5-bis(4-nitrophenyl) penta-1,4-dien-3-one on Trypanosoma cruzi and Its Combinational Effect with Benznidazole, Ketoconazole, or Fluconazole. Biomed Res Int. 2017;2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Souza JM, Vieira TM, Candido AC, Tezuka DY, Rao GS, de Albuquerque S, Crotti AEM, Siqueira-Neto JL, Magalhaes, LG. In vitro anti-Trypanosoma cruzi activity enhancement of curcumin by its monoketone tetramethoxy analog diveratralacetone. Curr Res Parasit Vector-Borne Dis. 2021;1(100031). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Polaquini CR, Morão LG, Nazaré AC, Torrezan GS, Dilarri G, Cavalca LB, Campos DL, Silva IC, Pereira JA, Scheffers DJ, Duque C, Pavan FR, Ferreira H, Regasini LO. Antibacterial activity of 3,3’-dihydroxycurcumin (DHC) is associated with membrane perturbation. Bioorg Chem. 2019;90: 103031. [DOI] [PubMed] [Google Scholar]
  • 54.Vieira TM, Dos Santos IA, Silva TS, Martins CHG, Crotti AEM. Antimicrobial Activity of Monoketone Curcuminoids Against Cariogenic Bacteria. Chem Biodivers. 2018;15(8): e1800216. [DOI] [PubMed] [Google Scholar]
  • 55.Nalli M, Ortar G, Schiano Moriello A, Di Marzo V, De Petrocellis L. Effects of curcumin and curcumin analogues on TRP channels. Fitoterapia. 2017;122: 126–131. [DOI] [PubMed] [Google Scholar]
  • 56.Badoco FR, Paula LAL, Orenha RP, Mendes TM, Squarisi IS, El-Sakkary N, Loiola MC, Katz N, Tavares DC, Sairre MI, Parreira RLT, Cabral FJ, Alegretti SM, Caffrey CR, Magalhaes LG. EF24, a schistosomicidal curcumin analog: Insights from its synthesis and phenotypic, biochemical and cytotoxic activities. Chem Biol Interact. 2022;368: 110191. [DOI] [PubMed] [Google Scholar]
  • 57.Torrezan G, Polaquini C, Lima M, Regasini L. Use of glycerol, waste glycerol from biodiesel production and other protic solvents in bioactive alpha, beta-unsaturated ketones synthesis. Sustain Chem Pharm. 2020;16. [Google Scholar]
  • 58.Polaquini CR, Marques BC, Ayusso GM, Morao LG, Sardi JCO, Campos DL, Silva IC, Cavalca LB, SCheffers DJ, Rosalen PL, Pavan FR, Ferreira H, Regasini LO. Antibacterial activity of a new monocarbonyl analog of curcumin MAC 4 is associated with divisome disruption. Bioorg Chem. 2021;109: 104668. [DOI] [PubMed] [Google Scholar]
  • 59.Monti L, Wang SC, Oukoloff K, Smith AB, Brunden KR, Caffrey CR, Ballatore C. Brain-Penetrant Triazolopyrimidine and Phenylpyrimidine Microtubule Stabilizers as Potential Leads to Treat Human African Trypanosomiasis. ChemMedChem. 2018;13(17): 1751–1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Faria J, Moraes CB, Song R, Pascoalini BS, Lee N, Siqueira-Neto JL, Cruz DJM, Parkinson T, Ioset JR, Cordeiro-da-Silva A, Freitas-Junior LH. Drug discovery for human African trypanosomiasis: identification of novel scaffolds by the newly developed HTS SYBR Green assay for Trypanosoma brucei. J Biomol Screen. 2015;20(1): 70–81. [DOI] [PubMed] [Google Scholar]
  • 61.O’Brien J, Wilson I, Orton T, Pognan F. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem. 2000;267(17): 5421–5426. [DOI] [PubMed] [Google Scholar]
  • 62.Changtam C, de Koning HP, Ibrahim H, Sajid MS, Gould MK, Suksamrarn A. Curcuminoid analogs with potent activity against Trypanosoma and Leishmania species. Eur J Med Chem. 2010;45(3): 941–956. [DOI] [PubMed] [Google Scholar]
  • 63.Alkhaldi AA, Creek DJ, Ibrahim H, Kim DH, Quashie NB, Burgess KE, Changtam C, Barrett MP, Suksamrarn A, de Koning HP. Potent trypanocidal curcumin analogs bearing a monoenone linker motif act on trypanosoma brucei by forming an adduct with trypanothione. Mol Pharmacol. 2015;87(3): 451–464. [DOI] [PubMed] [Google Scholar]
  • 64.Ettari R, Previti S, Tamborini L, Cullia G, Grasso S, Zappalà M. The Inhibition of Cysteine Proteases Rhodesain and TbCatB: A Valuable Approach to Treat Human African Trypanosomiasis. Mini Rev Med Chem. 2016;16(17): 1374–1391. [DOI] [PubMed] [Google Scholar]
  • 65.Steverding D, Caffrey CR. Should the enzyme name ‘rhodesain’ be discontinued? Mol Biochem Parasitol. 2021;245: 111395. [DOI] [PubMed] [Google Scholar]
  • 66.Ettari R, Previti S, Maiorana S, Allega A, Schirmeister T, Grasso S, Zappala M. Drug combination studies of curcumin and genistein against rhodesain of Trypanosoma brucei rhodesiense. Nat Prod Res. 2019;33(24): 3577–3581. [DOI] [PubMed] [Google Scholar]
  • 67.Jin F, Jin X, Jin Y, Sohn DW, Kim SA, Sohn DW, Kim YC, Kim HS. Structural requirements of 2’, 4’, 6’-tris(methoxymethoxy) chalcone derivatives for anti-inflammatory activity: The importance of a 2’-hydroxy moiety. Arch Pharm Res. 2007;30(11): 1359–1367. [DOI] [PubMed] [Google Scholar]
  • 68.Patterson S, Wyllie S. Nitro drugs for the treatment of trypanosomatid diseases: past, present, and future prospects. Trends Parasitol. 2014;30(6): 289–298. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting info

RESOURCES