Next Article in Journal
Asialo-rhuEPO as a Potential Neuroprotectant for Ischemic Stroke Treatment
Next Article in Special Issue
High Selectivity of 8-Hydroxyquinoline on Leishmania (Leishmania) and Leishmania (Viannia) Species Correlates with a Potent Therapeutic Activity In Vivo
Previous Article in Journal
Design of Potent Inhibitors Targeting the Main Protease of SARS-CoV-2 Using QSAR Modeling, Molecular Docking, and Molecular Dynamics Simulations
Previous Article in Special Issue
Antidiarrheal Potential of Viola canescens: In Vivo and In Silico Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Curcumin and Its Derivatives as Potential Antimalarial and Anti-Inflammatory Agents: A Review on Structure–Activity Relationship and Mechanism of Action

by
Siti Nur Hidayah Jamil
1,
Amatul Hamizah Ali
1,
Shevin Rizal Feroz
2,
Su Datt Lam
3,
Hani Kartini Agustar
4,
Mohd Ridzuan Mohd Abd Razak
5 and
Jalifah Latip
1,*
1
Department of Chemical Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
2
Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
3
Department of Applied Physics, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
4
Department of Earth Sciences and Environment, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
5
Herbal Medicine Research Centre, Institute for Medical Research, National Institute of Health (NIH) Complex, Ministry of Health Malaysia, Shah Alam 40170, Selangor, Malaysia
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(4), 609; https://doi.org/10.3390/ph16040609
Submission received: 20 February 2023 / Revised: 31 March 2023 / Accepted: 6 April 2023 / Published: 18 April 2023
(This article belongs to the Special Issue Natural Products for Treatment of Parasitic Diseases)

Abstract

:
Curcumin, one of the major ingredients of turmeric (Curcuma longa), has been widely reported for its diverse bioactivities, including against malaria and inflammatory-related diseases. However, curcumin’s low bioavailability limits its potential as an antimalarial and anti-inflammatory agent. Therefore, research on the design and synthesis of novel curcumin derivatives is being actively pursued to improve the pharmacokinetic profile and efficacy of curcumin. This review discusses the antimalarial and anti-inflammatory activities and the structure–activity relationship (SAR), as well as the mechanisms of action of curcumin and its derivatives in malarial treatment. This review provides information on the identification of the methoxy phenyl group responsible for the antimalarial activity and the potential sites and functional groups of curcumin for structural modification to improve its antimalarial and anti-inflammatory actions, as well as potential molecular targets of curcumin derivatives in the context of malaria and inflammation.

Graphical Abstract

1. Introduction

Malaria is a highly life-threatening infectious disease caused by the transmission of five Plasmodium parasites infecting the human body, which are P. falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi. P. falciparum, identified as being the most common and feared species infecting humans, is transmitted into the circulatory system through the infective bite of female Anopheles mosquitoes. This triggers the development of sporozoites, which can rapidly infect liver cells within 30 min after a bite (Figure 1). After approximately five days, the sporozoites mature into merozoites, and upon release from the cell, will begin to invade erythrocytes. The merozoites then multiply within the erythrocytes, leading to the release of more invasive merozoites to infect other erythrocytes. At this point, malarial-associated symptoms, namely, fever, headaches, body aches, general malaise, and rigors, will become apparent [1]. At the same time, mature merozoites will continue their development into gametocytes (sexual-stage parasites), which will subsequently form new sporozoites within female Anopheles mosquitoes to complete the Plasmodium life cycle [2,3].
Recent statistical data from the World Health Organization (WHO) indicate that 1.7 billion cases of malaria have been recorded worldwide, which led to 10.6 million deaths within the previous 20 years (2000–2020) [5]. Plasmodium infection and the consequent inflammation can lead to advanced and fatal malaria if left untreated, including cerebral and severe malaria, and other complications such as cardiovascular diseases. With severe and cerebral malaria being leading causes of mortality, many studies are currently focused on developing effective antimalarial and anti-inflammatory treatments. In addition, therapeutic efficacy studies (TESs) have shown increasing drug resistance in malaria, where currently available antimalarial drugs such as artemisinin and chloroquine are no longer effective against malaria infection. In line with the primary goal of the Global Fund Strategy 2023–2028, sourcing and developing novel and potent inhibitors to combat drug resistance in malaria treatment is now a necessity, hence, the growing list of bioactive compounds, which are proven for their antimalarial activities (Figure 2) [2,3,5,6,7].
Curcumin, which has been proven to be a promising candidate as an antimalarial and anti-inflammatory agent, is a natural polyphenolic compound, originating from the rhizomatous perennial plant, turmeric [10,11]. Curcumin was first named and reported by Vogel and Pelletier as a compound isolated from Curcuma longa rhizomes and was identified as an “orange-yellow substance” [12]. In commercially available curcumin, curcumin is present as a mixture of three curcuminoids, namely, curcumin (77%), bisdemethoxycurcumin (3%), and demethoxycurcumin (17%) [13] (Figure 3). Due to its high abundance among curcuminoids, curcumin is more accessible for research and drug development compared to other naturally sourced bioactive compounds that are present in low concentrations in the source plants [2].
Curcumin is known for its diverse applications worldwide. Aside from its use as a therapeutic remedy such as in traditional medicine and as an antiseptic agent, it is a popular cooking ingredient in many Asian countries. Further, it is applied as a coloring agent in manufacturing, food and beverage, and cosmetic industries [14]. Curcumin, which has been proven to show no toxicity even at high doses [15,16], has been approved as a “Generally Recognized as Safe” (GRAS) compound by the US Food and Drug Administration (FDA) [13]. However, the low bioavailability of curcumin limits its clinical applications and presents challenges in developing curcumin into a potent antimalarial drug candidate due to its poor oral absorbability, low aqueous solubility, and rapid metabolism in the body [17].
This review identifies curcumin derivatives with reported antimalarial and anti-inflammatory properties, evaluates the key functional groups/sites responsible for the antimalarial and anti-inflammatory activities, and discusses the mechanisms of action of these compounds that are associated with their biological effects. Several approaches to improve the activity of curcumin, including through drug combination, adjuvant application, curcumin nanoencapsulation, and structural modifications, have been previously suggested [18]. A specific approach focusing on the structural modification of the parent curcumin to enhance its bioactivity as an antimalarial and anti-inflammatory agent based on structure–activity relationship (SAR) analysis will also be expounded in this review. In addition, the absorption, distribution, metabolism, and excretion (ADME) properties of the curcumin derivatives will also be reviewed to evaluate their pharmacological profiles. Thus, this work is expected to be helpful in understanding the potential enhancement of curcumin bioactivity in malaria and inflammatory-related diseases, as well as in elucidating a detailed molecular-level mechanism of action associated with malarial infection.

2. Antimalarial and Anti-Inflammatory Activities of Curcumin and Its Structural Modifications

Curcumin has been reported to possess multiple therapeutic effects, including anti-inflammatory [19,20,21,22], anti-plasmodial [16,23,24,25], antifungal [16,26,27], antibacterial [28,29,30], antioxidant [31,32,33], and antitumor [16,34,35,36] activities. An understanding of its chemical structure has led to the identification of the sites responsible for its bioactivity, which are primarily its two methoxy phenolic groups (Figure 4). The hydroxy group attached at the para position of the aromatic phenyl group contributes toward the stability of curcumin, as the electrons from the hydroxy group are delocalized into the aromatic ring. Dohutia et al. investigated the effect of substituting the hydroxy group of curcumin with O-acetyl and methoxy groups on its antimalarial activity (Figure 5). The IC50 values from the in vitro study showed that substitution with an O-acetyl group (IC50 = 2.34 µM) exhibited similar potency as curcumin (IC50 = 3.25 µM); however, the potency decreased markedly (IC50 = 7.86 µM) when substituting with a methoxy group [37]. This was thought to be due to the biodegradability of the acetyl group through ester bond cleavage by esterases, which regenerates the parent curcumin structure, in contrast to the bond attached to an alkyl group, which is not readily cleaved [38]. Hence, this finding revealed the importance of the unsubstituted phenol group since the modification of the site led to the loss of curcumin’s antimalarial activity [38,39].
The need to develop potent inhibitors of proteins involved in the pathogenesis of malaria pathways is currently at a crisis point due to the increasing resistance of Plasmodium parasites against present antimalarial drugs and the lack of lead compounds for new antimalarial and anti-inflammatory agents. Thus, one approach that is actively implemented for malaria treatment is drug combination therapy [5,40,41,42,43]. Such an approach for the treatment of various diseases has long been applied, for example, to reduce drug resistance for tuberculosis in the 1960s [36]. According to the WHO, current front-line medications designed to mitigate the resistance of Plasmodium parasites against present antimalarial drugs are often based on artemisinin combination therapy (ACT), involving the combination of artemether–lumefantrine and amodiaquine (AL-AQ) [5]. Curcumin has also been used as a component in combination therapy against malaria. For instance, Tjahjani et al. showed that curcumin produced a synergistic antimalarial effect when administered together with dihydroartemisinin [44]. Furthermore, the combination of curcumin and piperine, an active compound found in black pepper, has proven to significantly enhance the bioavailability of curcumin by 2000% compared to curcumin alone [41].
Although drug combination therapy is proven for its efficacy, a potential drawback to its application are side effects, which are more likely compared to single-drug therapy [42]. Therefore, drug discovery by way of structural modification of existing bioactive compounds remains an important approach to combat drug resistance, along with drug combination therapy. The bioactivity of curcumin against the infected host and parasite target proteins is highly dependent on its reactivity [45,46]. However, the chemical structure of curcumin limits its bioavailability and stability at physiological pH and, thus, its efficacy as an antimalarial and anti-inflammatory agent [15,40]. Factors such as poor absorption, rapid metabolism, and rapid elimination contribute to the low bioavailability of curcumin in the body [15,47,48,49]. Therefore, deriving compounds with better pharmacokinetic properties than the parent curcumin is important in developing compounds with enhanced anti-inflammatory and antimalarial activities.
The instability and low bioavailability of curcumin are mainly due to the presence of the dicarbonyl group with a highly acidic α-H, which can easily resonate between keto and enolate forms, as illustrated in Figure 6 [50]. Previous research has indicated that the extremely reactive β-diketone group appears to be highly unstable and can be quickly degraded. Hence, curcumin derivatives are being actively studied to overcome the pharmacological limitations due to the chemistry of curcumin (Figure 7). Examples of curcumin derivatives exhibiting reasonable and better efficacy against various diseases than the parent curcumin are shown in Figure 8. A common strategy in designing curcumin derivatives with enhanced bioactivity is by lowering rotational flexibility and producing a more rigid orientation by reducing the reactivity of methylene alpha-H of the β-diketone moiety [51,52].
Before structural modification of curcumin is performed, the potential enhancement of curcumin bioactivity can be preliminarily suggested through pharmacokinetic analysis. Pharmacokinetic analysis is useful to predict the absorption, distribution, metabolism, and excretion (ADME) profiles of a compound and, thus, can provide supplementary screening data to evaluate the possible effects of a compound upon administration into the body.
The Swiss ADME server was employed to evaluate the potential of curcumin derivatives over the parent curcumin as drug candidates with enhanced antimalarial activity [62]. Table 1 shows the results of the analysis, which includes the predictions of physicochemical properties, drug-likeness according to Lipinski’s Rule of Five, and pharmacokinetic parameters for curcumin and its derivatives, as illustrated in Figure 9. The data also provide an overview of the ability of the compounds to form hydrogen bonds, which can be indicators of favorable interaction with protein active sites.
The majority of curcumin derivatives illustrated in Figure 9 are predicted to have better solubility, better GI absorbability, and BBB permeability than the parent curcumin. Enhanced interactions with the active site of proteins would theoretically delay the metabolism and elimination of a drug, hence, prolonging its duration of action. This analysis provides information to understand the possible enhancement of bioactivity contributed by structurally modifying the parent curcumin, which will be discussed in more detail in the later parts of this review.

3. Structure–Activity Relationship of Curcumin Derivatives as Antimalarial and Anti-Inflammatory Agents

The presence of various functional groups can influence the pharmacodynamic and pharmacokinetic effects of a drug. The unsubstituted methoxy phenol group of curcumin has been identified as crucial for its antimalarial and anti-inflammatory activity. Hence, the structural modification of curcumin is mostly focused on the β -dicarbonyl group and the highly acidic methylene α -hydrogen bridging the dicarbonyl groups.
In a study by Mishra et al., the keto–enol moiety was converted into the more rigid five-membered isoxazole and pyrazole rings, targeting the reactive methylene α -hydrogen [38,63]. The isoxazole and pyrazole derivatives of curcumin were synthesized based on the previously reported reaction schemes, as illustrated in Figure 10 [38,64,65]. The isoxazole and pyrazole derivatives have been reported for their improved anti-inflammatory and antiplasmodial effects, with the latter based on in vitro assays against both chloroquine-sensitive and chloroquine-resistant P. falciparum [38] (Table 2).
Interestingly, the isoxazole derivative (2) exhibited reduced antiplasmodial potency compared to the parent curcumin. Changing the oxygen atom of the isoxazole ring into a nitrogen atom, thus, forming the pyrazole derivative (3) (Figure 11), greatly enhanced the activity, with an IC50 value of 0.48 μ M, compared to 3.25 μ M of the parent curcumin. Thus, the presence of nitrogen is vital for the antimalarial activity of the curcumin derivative.
The activity of the pyrazole derivatives (48) upon the addition of a phenyl group to the nitrogen atom was also investigated (Figure 12). The formation of derivatives (5), (6), and (7) revealed that the presence of different functional groups on the phenyl ring greatly affects the potency of the compounds. Derivative (5), which contains fluorine as opposed to chlorine, as in derivative (7), showed significantly higher activity, as can be seen in Table 2. This can be attributed to the lower steric hindrance caused by fluorine compared to chlorine, leading to a more favorable structural characteristic for interaction with other molecules. In addition, the low IC50 value of derivative (6), which contains a nitro group attached to the phenyl (Figure 13), indicates that the electron withdrawing group enhances the activity and potency of curcumin derivatives against malaria. This is believed to be due to the negative mesomeric effect, which delocalized and withdrew electrons from the ring, hence stabilizing the compound [66].
Similar to derivatives synthesized by the Knoevenagel reaction (9, 10, 11, and 1442), the presence of an electron donating group (8) resulted in a negative effect on the antimalarial activity of the derivative, with lower potency compared to the parent curcumin. Therefore, these observations demonstrate that electron-withdrawing substituents are crucial to enhancing the antimalarial activity of curcumin derivatives.
Apart from the pyrazole derivatives, a series of Knoevenagel condensate derivatives (911) were also derived from curcumin and tested for in vitro antimalarial activity by Mishra et al. [38,67]. The curcumin derivatives were then extendedly synthesized by Dohutia et al. (Table 3) (Figure 14) through further structural modifications producing compounds 1442, whose reactive methylene α -hydrogen was substituted with a phenyl group, as shown in Figure 15 [37]. It was suggested that the Knoevenagel condensate derivatives were able to inhibit NF- κ β [38,68] and PfATP6 [39,69] based on in silico studies, and exhibited varying levels of antimalarial and anti-inflammatory activities (Figure 15).
Parameters based on Lipinski’s rule of five, such as solubility and log P, as well as the number of H-bond donors and acceptors, were evaluated in the study. These parameters provide information on the stability and bioavailability of curcumin derivatives and facilitate the design of the most potent derivative structures [71,72]. The Knoevenagel condensate derivatives produced generally exhibited enhanced antimalarial potential, based on their better solubility, binding energy, and percentage of schizont inhibition.
The addition of the phenyl group substituting the α -H (9) provides a steric effect and deactivates the reactive methylene site. The extended substituent at the α -carbon position also enables additional interactions with proteins through hydrophobic interactions, resulting in higher free binding energy and percentage of schizont inhibition. The enhanced solubility and log P value of the derivatives translate to their enhanced stability within the hydrophilic and hydrophobic regions of the binding sites on the target protein compared to the parent curcumin (1).
The functional groups introduced to the aromatic phenyl also affect the binding and percentage of schizont inhibition of the derivatives. An additional hydroxy group at the meta position (15) increases the binding energy through the introduction of additional H-bond donors and acceptors. The hydroxy group also limits the free rotation of the phenyl group, contributing to steric effect and improved interaction with protein, leading to higher free binding energy and percentage of schizont inhibition than derivative (9) [73].
Interestingly, the substitution of the meta hydroxy with a methoxy group combined with the addition of another hydroxy at the para position (11) further enhanced the potency of the compound compared to both curcumin (1) and derivative (9) (Table 2). However, eliminating the methoxy group at the meta position, leaving a monosubstituted para-hydroxy phenol group (10), had the opposite effect of reducing its potency to a level lower than even the parent curcumin (Table 2). Thus, the presence of both the electron-donating methoxy group at the meta position and the electron-withdrawing hydroxy group is crucial for the improved antimalarial and anti-inflammatory activities of the curcumin derivative (Figure 16).
Comparing the Knoevenagel condensate derivatives, compound (21), which has fluorine (EWG) attached to the phenyl (Figure 17), exhibited the highest percentage of schizont inhibition and the strongest binding. The highly negative and electron-withdrawing fluorine confers structural stability through an inductive effect, similar to the hydroxyl group.
Another approach to eliminating the keto–enol and the active methylene moieties is through the reduction of the dicarbonyl group into a monocarbonyl group. The compounds reported by Yusuf et al. [74] were not synthesized by modification of the parent curcumin, but instead by total synthesis, as shown in Figure 18. The bioactivity of each monocarbonyl derivative against P. falciparum was assessed based on an in silico study of their interaction with the PfDXR protein, as well as an in vitro antimalarial assay, initially developed by Hanne et al. and Mukhtar et al. [74,75,76]. Notably, the activity of the monocarbonyl derivatives was observed to be highly dependent on the substituents attached to the aromatic phenyl rings [74].
The mechanism of action for this range of curcumin derivatives was identified to be through the inhibition of the parasite protein PfDXR, which is a proven target for the antimalarial drug fosmidomycin [77]. The carbonyl group at the center of the compounds facilitates a ligand–protein interaction through the formation of two hydrogen bonds at the center of the binding pocket. The hydrophobic aromatic region of the derivatives also contributes to additional hydrophobic interactions, which were not present with fosmidomycin.
Monocarbonyl derivative (43), which maintained the methoxy phenol group present in the parent curcumin (1) (Figure 19), showed 55% parasite elimination in the study. Upon further modification by substituting the methoxy and hydroxy groups into a monosubstituted ring, derivatives (44, 45) were produced. However, the percentage of parasite elimination decreased, indicating their reduced potency. This result supports the importance of the unsubstituted methoxy phenol group of curcumin, as modifying this site leads to the loss of its antimalarial activity [38,39].

4. Mechanism of Action of Curcumin

Finding a safe and effective anti-inflammatory therapy remains an obstacle in antimalarial drug development. Malarial infection affects the regulation of the immune system and inflammation levels within the body. Curcumin and its derivatives have emerged as an attractive anti-inflammatory agent due to their wide range of effective cellular-level actions, including regulating the levels of transcription factors [78], cytokine expression [78], and enzymes involved in the progression of infected cells [40,61].

4.1. Host Proteins as Molecular Targets of Curcumin

Since malaria causes dysregulation in the inflammatory response, the elucidation of the mechanisms of action of malarial infection is important in order to understand the pathways and binding targets for inhibition. One potential mechanism involved in regulating the pathophysiology of malaria was identified to involve the Toll-like receptor (TLR) signaling pathways. TLRs, which are located on the cell surface, recognize the released Plasmodium DNA and trigger the initiation of immune responses through the activation of NF- κ B [79,80]. The transcription factor NF- κ B is a critical signaling protein involved in various inflammatory responses and gene expressions [68,81,82]. NF- κ B is found in a dormant state in the cytoplasm and will only be transcribed when it is activated and translocated into the nucleus [80,83]. This transcription factor is the main target protein that directly regulates the pro- and anti-inflammatory cytokines within the body, including COX-2, TNF, IL-1, IL-6, IL-8, IL-10, and chemokines [79,84,85,86]. The dysregulation of these cytokines and chemokines will lead to the expression of malarial symptoms such as fever, and if left untreated, will lead to severe malaria (Figure 20). Therefore, NF- κ B has become the most targeted factor in the development of antimalarial agents. Another study also established and reported that P. falciparum infection leads to the elevation of TNF production, which is the main malarial pathogenic factor, and, hence, could lead to a high risk of severe malaria and even death [87,88,89].
Curcumin has been proven to suppress the activation and translocation of NF- κ B into the nucleus, thus, controlling the level of inflammatory cytokines and the larger inflammatory response. Its ability to interact and inhibit various proteins helps in the elucidation and modulation of the pathophysiology of diseases at the molecular level, including malaria [8,90,91]. The interaction of curcumin with proteins is facilitated by its structural flexibility conferred by the presence of the unsaturated diketo group at the center of curcumin [8,85] (Figure 21).
A recent study by Ali et al. on TLR pathways involving the control of the protein kinases Akt and glycogen synthase kinase-3 β (GSK3 β ) also proved NF- κ B as a downstream target that regulates anti-inflammatory cytokines [92]. Based on in vivo studies, curcumin was demonstrated to directly inhibit the host GSK3 β , leading to the phosphorylation of NF- κ B, hence, modulating the regulation of pro- (TNF- α , IFN- γ , and IL-18) and anti-inflammatory (IL-4 and IL-10) cytokine levels [92]. The immunomodulating effect of curcumin in reducing pro-inflammatory cytokine expression can potentially prevent severe and cerebral malaria [93,94]. As evidence to this claim, several in vitro studies have shown that curcumin downregulates pro-inflammatory cytokine production and the expression of cell adhesion molecules in TNF-activated human endothelial cells observed at the trophozoites stage of P. falciparum transmission [93]. Furthermore, the inhibition of NF- κ B by curcumin also suppresses the generation of reactive oxygen species (ROS), which attenuates the inflammatory response [95,96,97].

4.2. Parasite Proteins as Molecular Targets of Curcumin

With regard to parasite proteins, curcumin induces ROS generation in parasite cells, which affects the function of the PfGCN5 histone acetyltransferases (HATs) p300/CREB-binding protein (CBP), thus, inhibiting histone acetylation and the transcription process in the parasite [98]. The generation of ROS is an important antimalarial mechanism as it induces protein and DNA damage within parasite cells, leading to their death [92,99,100]. The specific inhibition of parasite HAT by curcumin prevents the acetylation of K9 and K14 of histone H3. Cui et al. also reported that the antiplasmodial activity of curcumin is attributed, at least in part, to the production of ROS and the downregulation of PfGCN5 HAT activity [48].
Another mechanism of action of curcumin is by disrupting the transmission and development of Plasmodium parasites at the erythrocytic stage. As erythrocytes burst to release more merozoites, heme is also released. However, the released heme is highly toxic to the merozoites. Thus, the parasites will be stimulated to convert hematin into its detoxified polymeric form, hemozoin [3,47,101]. Curcumin treatment was reported to inhibit the formation of hemozoins in vitro, as observed through transmission electron microscopy in a study using the P. falciparum 3D7 strain [44]. This proved that the antimalarial activity of curcumin is similar to that of quinine and chloroquine [101].
Uncontrolled parasite transmission in the body can develop into fatal severe anemia or cerebral malaria, whose pathophysiology involves the inflammatory response [91,102]. The excessive stimulation of pro-inflammatory cytokines by the parasite subsequently leads to the sequestration of parasites in the brain [103]. Several reports have indicated the ability of curcumin to eliminate parasites at the trophozoite stage, synergistically and effectively better than artemisinin [37,38,63,104]. This observation was also proven using chloroquine-sensitive (CQS) and chloroquine-resistant (CQR) P. falciparum strains, with a proposed mechanism of curcumin action against the parasite proteins PfRIO2-kinase and PfGCN5 HAT [38,104,105].
The Knoevenagel condensate curcumin derivatives mentioned earlier were suggested to target the PfATP6 parasite protein to explain their schizont inhibition activity. This was based on the established target of the reference antimalarial drug, artemisinin, which shares the same binding pocket on PfATP6 as curcumin [37,69]. The study applied PreADMET predictions, which demonstrated the attachment and interaction of curcumin and its derivatives with the active site of PfATP6.
The identification of therapeutic targets involved in malarial infection can provide a better understanding of the inhibitory mechanism of antimalarial agents (Figure 22) [39,106]. The current knowledge on the action of curcumin derivatives is limited, as their host-targeting mechanisms have not been entirely established (Table 4) [41,90,107]. Therefore, future research that can explain an in-depth understanding of the molecular-level mechanism of action of curcumin and its bioactive derivatives will not only help in the development of potent antimalarial and anti-inflammatory agents [108] but also for other diseases [109,110,111].

5. Conclusions

Various therapeutic effects of curcumin against a wide range of diseases have been extensively reported. Within the context of malarial infection, curcumin exhibits the dual antiplasmodial activity against parasites as well as anti-inflammatory action within the host. However, due to its limited bioavailability, the synthesis of derivatives with better pharmacological profiles than the parent curcumin through structural modifications has been explored. The incorporation of pharmacokinetic property prediction and structure–activity relationship data to rationalize the design of curcumin derivatives with enhanced bioactivity paves the way for the development of more potent antimalarial medicine. While preserving the methoxy phenol groups, which are crucial for the antimalarial and anti-inflammatory activities of curcumin, the reactive methylene group bridging the two carbonyl groups has been targeted for structural modification to generate promising curcumin derivatives. The presence of electron-withdrawing and electron-donating groups within the curcumin derivatives significantly affects their potency, pharmacokinetics, and physicochemical properties, as evaluated in terms of free binding energy, solubility, lipophilicity, IC50 values, and percentage of schizont inhibition. Even though the pharmacological effects of curcumin have been attributed to the inhibition of multiple signaling pathways and enzymes in various biological systems, the detailed and specific molecular mechanism underlying its parasiticidal and anti-inflammatory activities remains to be elucidated in detail. The identification of specific target proteins for inhibition that correlates with the mechanism of action of curcumin and its various derivatives is essential for developing safer and more effective therapies against malaria.

Author Contributions

Conceptualization, S.N.H.J., A.H.A., S.R.F., S.D.L., H.K.A., M.R.M.A.R. and J.L.; methodology, S.N.H.J. and A.H.A.; writing—original draft preparation, S.N.H.J.; writing—review and editing, A.H.A., S.R.F., S.D.L., J.L., H.K.A. and M.R.M.A.R.; supervision, S.R.F., S.D.L. and J.L.; funding acquisition, J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by UNIVERSITI KEBANGSAAN MALAYSIA (grant no: GUP-2021-047).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

The authors express deep gratitude for the student financial assistance provided by the UKM Zamalah Research Scheme throughout the research and writing of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Centers for Disease Control and Prevention (CDC), 2022. Available online: https://www.cdc.gov/malaria (accessed on 21 March 2023).
  2. Habibi, P.; Grossi-de-Sa, M.F.; Shi, Y.; Khan, I. Plants as sources of natural and recombinant antimalaria agents. Mol. Biotechnol. 2022, 64, 1177–1197. [Google Scholar] [CrossRef] [PubMed]
  3. Kingston, D.G.I.; Cassera, M.B. Antimalarial natural products. Prog. Chem. Org. Nat. Prod. 2022, 117, 1–106. [Google Scholar] [PubMed]
  4. Moreno-García, M.; Recio-Tótoro, B.; Claudio-Piedras, F.; Lanz-Mendoza, H. Injury and immune response: Applying the danger theory to mosquitoes. Front. Plant Sci. 2014, 5, 451. [Google Scholar] [CrossRef] [PubMed]
  5. WHO, World Health Organisation. World Malaria Report, Geneva, 2022. Available online: https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2022 (accessed on 21 March 2023).
  6. Ridley, R.G. Medical need, scientific opportunity and the drive for antimalarial drugs. Nature 2002, 415, 686–693. [Google Scholar] [CrossRef] [PubMed]
  7. Greenwood, B.; Mutabingwa, T. Malaria in 2002. Nature 2002, 415, 670–672. [Google Scholar] [CrossRef]
  8. Dasgupta, T.; Chitnumsub, P.; Kamchonwongpaisan, S.; Maneeruttanarungroj, C.; Swift, S.E.; Lyons, T.M.; Tirado-Rives, J.; Jorgensen, W.L.; Yuthavong, Y.; Anderson, K.S. Exploiting structural analysis, in silico screening, and serendipity to identify novel inhibitors of drug-resistant falciparum malaria. ACS Chem. Biol. 2009, 4, 29–40. [Google Scholar] [CrossRef]
  9. Alson, S.G.; Jansen, O.; Cieckiewicz, E.; Rakotoarimanana, H.; Rafatro, H.; Degotte, G.; Francotte, P.; Frederich, M. In-vitro and in-vivo antimalarial activity of caffeic acid and some of its derivatives. J. Pharm. Pharmacol. 2018, 70, 1349–1356. [Google Scholar] [CrossRef]
  10. Urošević, M.; Nikolić, L.; Gajić, I.; Nikolić, V.; Dinić, A.; Miljković, V. Curcumin: Biological activities and modern pharmaceutical forms. Antibiotics 2022, 11, 135. [Google Scholar] [CrossRef]
  11. Cheng, A.L.; Hsu, C.H.; Lin, J.K.; Hsu, M.M.; Ho, Y.F.; Shen, T.S. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 2001, 21, 2895–2900. [Google Scholar]
  12. Aggarwal, B.B.; Bhatt, I.D.; Ichikawa, H.; Ahn, K.S.; Sethi, G.; Sandur, S.K.; Natarajan, C.; Seeram, N.; Shishodia, S. 10 Curcumin—Biological and Medicinal Properties; Indsaff, Inc.: Batala, India, 2006; p. 297. [Google Scholar]
  13. Jayaprakasha, G.K.; Rao, L.J.; Sakariah, K.K. Antioxidant activities of curcumin, demethoxycurcumin and bisdemethoxycurcumin. Food Chem. 2006, 98, 720–724. [Google Scholar] [CrossRef]
  14. Hewlings, S.J.; Kalman, D.S. Curcumin: A review of its effects on human health. Foods 2017, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  15. Basnet, P.; Skalko-Basnet, N. Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 2011, 16, 4567–4598. [Google Scholar] [CrossRef] [PubMed]
  16. Hatcher, H.; Planalp, R.; Cho, J.; Torti, F.M.; Torti, S.V. Curcumin: From ancient medicine to current clinical trials. Cell. Mol. Life Sci. 2008, 65, 1631–1652. [Google Scholar] [CrossRef] [PubMed]
  17. Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef]
  18. Oglah, M.K.; Mustafa, Y.F.; Bashir, M.K.; Jasim, M.H.; Mustafa, Y.F. Curcumin and its derivatives: A review of their biological activities. Syst. Rev. Pharm. 2020, 11, 472. [Google Scholar]
  19. Lee, K.H.; Aziz, F.H.A.; Syahida, A.; Abas, F.; Shaari, K.; Israf, D.A.; Lajis, N.H. Synthesis and biological evaluation of curcumin-like diarylpentanoid analogues for anti-inflammatory, antioxidant and anti-tyrosinase activities. Eur. J. Med. Chem. 2009, 44, 3195–3200. [Google Scholar] [CrossRef]
  20. Liang, G.; Yang, S.; Jiang, L.; Zhao, Y.; Shao, L.; Xiao, J.; Ye, F.; Li, Y.; Li, X. Synthesis and anti-bacterial properties of mono-carbonyl analogues of curcumin. Chem. Pharm. Bull. 2008, 56, 162–167. [Google Scholar] [CrossRef]
  21. Dai, C.; Ciccotosto, G.D.; Cappai, R.; Tang, S.; Li, D.; Xie, S.; Xiao, X.; Velkov, T. Curcumin attenuates colistin-induced neurotoxicity in N2a cells via anti-inflammatory activity, suppression of oxidative stress, and apoptosis. Mol. Neurobiol. 2018, 55, 421–434. [Google Scholar] [CrossRef]
  22. Yuan, J.; Liu, R.; Ma, Y.; Zhang, Z.; Xie, Z. Curcumin attenuates airway inflammation and airway remolding by inhibiting NF-κB signaling and COX-2 in cigarette smoke-induced COPD mice. Inflammation 2018, 41, 1804–1814. [Google Scholar] [CrossRef]
  23. Nandakumar, D.N.; Nagaraj, V.A.; Vathsala, P.G.; Rangarajan, P.; Padmanaban, G. Curcumin-artemisinin combination therapy for malaria. Antimicrob. Agents Chemother. 2006, 50, 1859–1860. [Google Scholar] [CrossRef]
  24. Martinelli, A.; Rodrigues, L.A.; Cravo, P. Plasmodium chabaudi: Efficacy of artemisinin+ curcumin combination treatment on a clone selected for artemisinin resistance in mice. Exp. Parasitol. 2008, 119, 304–307. [Google Scholar] [CrossRef] [PubMed]
  25. Reddy, R.C.; Vatsala, P.G.; Keshamouni, V.G.; Padmanaban, G.; Rangarajan, P.N. Curcumin for malaria therapy. Biochem. Biophys. Res. Commun. 2004, 326, 472–474. [Google Scholar] [CrossRef] [PubMed]
  26. Dovigo, L.N.; Pavarina, A.C.; Ribeiro, A.P.D.; Brunetti, I.L.; Costa, C.A.D.S.; Jacomassi, D.P.; Bagnato, V.S.; Kurachi, C. Investigation of the photodynamic effects of curcumin against Candida albicans. Photochem. Photobiol. 2011, 87, 895–903. [Google Scholar] [CrossRef] [PubMed]
  27. Garcia-Gomes, A.S.; Curvelo, J.A.R.; Soares, R.A.; Ferreira-Pereira, A. Curcumin acts synergistically with fluconazole to sensitize a clinical isolate of Candida albicans showing a MDR phenotype. Med. Mycol. J. 2012, 50, 26–32. [Google Scholar] [CrossRef] [PubMed]
  28. Tong, S.Y.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef]
  29. Gunes, H.; Gulen, D.; Mutlu, R.; Gumus, A.; Tas, T.; Topkaya, A.E. Antibacterial effects of curcumin: An in vitro minimum inhibitory concentration study. Toxicol. Ind. Health 2016, 32, 246–250. [Google Scholar] [CrossRef]
  30. Khudhayer, O.M.; Fakri Mustafa, Y. Curcumin analogs: Synthesis and biological activities. Med. Chem. Res. 2020, 29, 479–486. [Google Scholar] [CrossRef]
  31. Jagetia, G.C.; Rajanikant, G.K. Curcumin stimulates the antioxidant mechanisms in mouse skin exposed to fractionated γ-irradiation. Antioxidants 2015, 4, 25–41. [Google Scholar] [CrossRef]
  32. Ma, Q.; Ren, Y.; Wang, L. Investigation of antioxidant activity and release kinetics of curcumin from tara gum/polyvinyl alcohol active film. Food Hydrocoll. 2017, 70, 286–292. [Google Scholar] [CrossRef]
  33. Zhu, Q.; Sun, Y.; Yun, X.; Ou, Y.; Zhang, W.; Li, J.X. Antinociceptive effects of curcumin in a rat model of postoperative pain. Sci. Rep. 2014, 4, 1–4. [Google Scholar] [CrossRef]
  34. Kunnumakkara, A.B.; Anand, P.; Aggarwal, B.B. Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett. 2008, 269, 199–225. [Google Scholar] [CrossRef] [PubMed]
  35. Shao, Z.M.; Shen, Z.Z.; Liu, C.H.; Sartippour, M.R.; Go, V.L.; Heber, D.; Nguyen, M. Curcumin exerts multiple suppressive effects on human breast carcinoma cells. Int. J. Cancer 2002, 98, 234–240. [Google Scholar] [CrossRef] [PubMed]
  36. Momtazi-Borojeni, A.A.; Mosafer, J.; Nikfar, B.; Ekhlasi-Hundrieser, M.; Chaichian, S.; Mehdizadehkashi, A.; Vaezi, A. Curcumin in advancing treatment for gynecological cancers with developed drug-and radiotherapy-associated resistance. Rev. Physiol. Biochem. Pharmacol. 2019, 176, 107–129. [Google Scholar]
  37. Dohutia, C.; Chetia, D.; Gogoi, K.; Bhattacharyya, D.R.; Sarma, K. Molecular docking, synthesis and in vitro antimalarial evaluation of certain novel curcumin analogues. Braz. J. Pharm. Sci. 2018, 53, 1–14. [Google Scholar] [CrossRef]
  38. Mishra, S.; Karmodiya, K.; Surolia, N.; Surolia, A. Synthesis and exploration of novel curcumin analogues as anti-malarial agents. Bioorg. Med. Chem. 2008, 16, 2894–2902. [Google Scholar] [CrossRef] [PubMed]
  39. Eckstein-Ludwig, U.; Webb, R.J.; Van Goethem, I.D.A.; East, J.M.; Lee, A.G.; Kimura, M.; O’neill, P.M.; Bray, P.G.; Ward, S.A.; Krishna, S. Artemisinins target the SERCA of Plasmodium falciparum. Nature 2003, 424, 957–961. [Google Scholar] [CrossRef]
  40. Hosseini-Zare, M.S.; Sarhadi, M.; Zarei, M.; Thilagavathi, R.; Selvam, C. Synergistic effects of curcumin and its analogs with other bioactive compounds: A comprehensive review. Eur. J. Med. Chem. 2021, 210, 113072. [Google Scholar] [CrossRef]
  41. Khairani, S.; Fauziah, N.; Wiraswati, H.L.; Panigoro, R.; Setyowati, E.Y.; Berbudi, A. The potential use of a curcumin-piperine combination as an antimalarial agent: A systematic review. J. Trop. Med. 2021, 2021, 9135617. [Google Scholar] [CrossRef]
  42. Mokhtari, R.B.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022. [Google Scholar] [CrossRef]
  43. Möhrle, J.J. Towards the next generation of antimalaria combination therapies. Lancet Infect. Dis. 2021, 21, 1620–1621. [Google Scholar] [CrossRef]
  44. Tjahjani, S.; Syafruddin; Tjokropranoto, R. Interaction of alphamangostin and curcumin with dihydroartemisinin as antimalaria in vitro. IOP Conf. Ser. Earth Environ. Sci. 2018, 125, 012017. [Google Scholar] [CrossRef]
  45. Aggarwal, B.B.; Harikumar, K.B. 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, 40–59. [Google Scholar] [CrossRef]
  46. 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, 2623–2631. [Google Scholar] [CrossRef] [PubMed]
  47. Hsu, C.H.; Cheng, A.L. Clinical studies with curcumin. Adv. Exp. Med. Biol. 2007, 595, 471–480. [Google Scholar] [PubMed]
  48. Pan, M.H.; Huang, T.M.; Lin, J.K. Biotransformation of curcumin through reduction and glucuronidation in mice. Drug Metab. Dispos. 1999, 27, 486–494. [Google Scholar] [PubMed]
  49. Sharma, R.A.; Steward, W.P.; Gescher, A.J. Pharmacokinetics and pharmacodynamics of curcumin. The molecular targets and therapeutic uses of curcumin in health and disease. AEMB 2007, 595, 453–470. [Google Scholar]
  50. da Silva, C.C.; Pacheco, B.S.; das Neves, R.N.; Alves, M.S.D.; Sena-Lopes, A.; Moura, S.; Borsuk, S.; de Pereira, C.M.P. Antiparasitic activity of synthetic curcumin monocarbonyl analogues against Trichomonas vaginalis. Biomed. Pharmacother. 2019, 111, 367–377. [Google Scholar] [CrossRef]
  51. Narlawar, R.; Pickhardt, M.; Leuchtenberger, S.; Baumann, K.; Krause, S.; Dyrks, T.; Weggen, S.; Mandelkow, E.; Schmidt, B. Curcumin-derived pyrazoles and isoxazoles: Swiss army knives or blunt tools for Alzheimer’s disease? ChemMedChem 2008, 3, 165–172. [Google Scholar] [CrossRef]
  52. Chakraborti, S.; Dhar, G.; Dwivedi, V.; Das, A.; Poddar, A.; Chakraborti, G.; Basu, G.; Chakrabarti, P.; Surolia, A.; Bhattacharyya, B. Stable and potent analogues derived from the modification of the dicarbonyl moiety of curcumin. Biochemistry 2013, 52, 7449–7460. [Google Scholar] [CrossRef]
  53. Zielińska, A.; Alves, H.; Marques, V.; Durazzo, A.; Lucarini, M.; Alves, T.F.; Morsink, M.; Willemen, N.; Eder, P.; Chaud, M.V.; et al. Properties, extraction methods, and delivery systems for curcumin as a natural source of beneficial health effects. Medicina 2020, 56, 336. [Google Scholar] [CrossRef]
  54. Angelini, G.; Pasc, A.; Gasbarri, C. Curcumin in silver nanoparticles aqueous solution: Kinetics of keto-enol tautomerism and effects on AgNPs. Colloids Surf. A Physicochem. Eng. Asp. 2020, 603, 125235. [Google Scholar] [CrossRef]
  55. Thomachan, S.; Sindhu, S.; John, V.D. Synthesis, characterization, antibacterial, antifungal and cytotoxic activity of curcuminoid analogues with trisubstituted phenyl and anthracenyl ring and their zinc (II), copper (II) and vanadyl (IV) chelates. Int. J. Pharmac. Chem. 2016, 6, 78–86. [Google Scholar]
  56. Al-Hujaily, E.M.; Mohamed, A.G.; Al-Sharif, I.; Youssef, K.M.; Manogaran, P.S.; Al-Otaibi, B.; Al-Haza’a, A.; Al-Jammaz, I.; Al-Hussein, K.; Aboussekhra, A. PAC, a novel curcumin analogue, has anti-breast cancer properties with higher efficiency on ER-negative cells. Breast Cancer Res. Treat. 2011, 128, 97–107. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, J.; Cai, Z.; Wei, X.; Chen, M.; Ying, S.; Shi, L.; Xu, R.A.; He, F.; Liang, G.; Zhang, X. Anti-lung cancer activity of the curcumin analog JZ534 in vitro. Biomed. Res. Int. 2015, 2015, 504529. [Google Scholar] [CrossRef]
  58. Tan, X.; Sidell, N.; Mancini, A.; Huang, R.P.; Wang, S.; Horowitz, I.R.; Liotta, D.C.; Taylor, R.N.; Wieser, F. Multiple anticancer activities of EF24, a novel curcumin analog, on human ovarian carcinoma cells. Reprod. Sci. 2010, 17, 931–940. [Google Scholar] [CrossRef]
  59. Pan, Y.; Wang, Y.; Cai, L.; Cai, Y.; Hu, J.; Yu, C.; Li, J.; Feng, Z.; Yang, S.; Li, X.; et al. Inhibition of high glucose-induced inflammatory response and macrophage infiltration by a novel curcumin derivative prevents renal injury in diabetic rats. Br. J. Pharmacol. 2012, 166, 1169–1182. [Google Scholar] [CrossRef]
  60. Katsori, A.M.; Chatzopoulou, M.; Dimas, K.; Kontogiorgis, C.; Patsilinakos, A.; Trangas, T.; Hadjipavlou-Litina, D. Curcumin analogues as possible anti-proliferative & anti-inflammatory agents. Eur. J. Med. Chem. 2011, 46, 2722–2735. [Google Scholar]
  61. Chougala, M.B.; Bhaskar, J.J.; Rajan, M.G.R.; Salimath, P.V. Effect of curcumin and quercetin on lysosomal enzyme activities in streptozotocin-induced diabetic rats. Clin. Nutr. 2012, 31, 749–755. [Google Scholar] [CrossRef]
  62. 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]
  63. Rodrigues, F.C.; Anil Kumar, N.V.; Thakur, G. The potency of heterocyclic curcumin analogues: An evidence-based review. Pharmacol. Res. 2021, 166, 105489. [Google Scholar] [CrossRef]
  64. Flynn, D.L.; Belliotti, T.R.; Boctor, A.M.; Connor, D.T.; Kostlan, C.R.; Nies, D.E.; Ortwine, D.F.; Schrier, D.J.; Sircar, J.C. Styrylpyrazoles, styrylisoxazoles, and styrylisothiazoles. Novel 5-lipoxygenase and cyclooxygenase inhibitors. J. Med. Chem. 1991, 34, 518–525. [Google Scholar] [CrossRef] [PubMed]
  65. Selvam, C.; Jachak, S.M.; Thilagavathi, R.; Chakraborti, A.K. Design, synthesis, biological evaluation and molecular docking of curcumin analogues as antioxidant, cyclooxygenase inhibitory and anti-inflammatory agents. Bioorganic Med. Chem. Lett. 2005, 15, 1793–1797. [Google Scholar] [CrossRef] [PubMed]
  66. Littlejohn, A.C.; Smith, J.W. The dipole moments of some aromatic nitro-compounds in relation to the steric inhibition of the mesomeric effect of the nitro-group. J. Chem. Soc. 1957, 1957, 2476–2482. [Google Scholar] [CrossRef]
  67. Jones, G. The Knoevenagel Condensation. Org. React. 2011, 15, 204–599. [Google Scholar]
  68. Zambre, A.P.; Kulkarni, V.M.; Padhye, S.; Sandur, S.K.; Aggarwal, B.B. Novel curcumin analogs targeting TNF-induced NF-κB proliferation in human leukemic KBM-5 cellsB activation. Bioorg. Med. Chem. 2006, 14, 7196–7204. [Google Scholar] [CrossRef] [PubMed]
  69. Ji, H.F.; Shen, L. Interactions of curcumin with the PfATP6 model and the implications for its antimalarial mechanism. Bioorganic Med. Chem. Lett. 2009, 19, 2453–2455. [Google Scholar] [CrossRef]
  70. NCBI, National Center for Biotechnology Information. PubChem Compound Summary for CID 969516, Curcumin. PubChem 2022. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/curcumin (accessed on 22 December 2022).
  71. Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of drug absorption using multivariate statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef]
  72. Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef]
  73. Harrold, M.W.; Zavod, R.M. Functional Group Characteristics and Roles in Basic Concepts in Medicinal Chemistry; American Society of Health-System Pharmacists Publications: Bethesda, MD, USA, 2018; pp. 22–66. [Google Scholar]
  74. Yusuf, A.S.; Sada, I.; Hassan, Y.; Olomola, T.O.; Adeyemi, C.M.; Ajibade, S.O. Synthesis, antimalarial activity, and docking studies of monocarbonyl analogues of curcumin. Ovidius Univ. Ann. Chem. 2018, 29, 92–96. [Google Scholar] [CrossRef]
  75. Ziegler, H.L.; Stærk, D.; Christensen, J.; Hviid, L.; Hagerstrand, H.; Jaroszewski, J.W. In vitro Plasmodium falciparum drug sensitivity assay: Inhibition of parasite growth by incorporation of stomatocytogenic amphiphiles into the erythrocyte membrane. Antimicrob. Agents Chemother. 2002, 46, 1441–1446. [Google Scholar] [CrossRef]
  76. Mukhtar, M.D.; Bashir, M.; Arzai, A.H. Comparative in-vitro studies on antiplasmodial quality of some Nigerian and foreign brands of chloroquine oral formulations marketed in kano. Afr. J. Biotechnol. 2006, 5, 2464. [Google Scholar]
  77. Bodill, T.; Conibear, A.C.; Blatch, G.L.; Lobb, K.A.; Kaye, P.T. Synthesis and evaluation of phosphonated N-heteroarylcarboxamides as DOXP-reductoisomerase (DXR) inhibitors. Bioorg. Med. Chem. 2011, 19, 1321–1327. [Google Scholar] [CrossRef] [PubMed]
  78. Ni, H.; Jin, W.; Yuan, B.; Zhu, T.; Wang, J.; Jiang, J.; Liang, W.; Ma, Z. Curcumin inhibits the increase of labile zinc and the expression of inflammatory cytokines after traumatic spinal cord injury in rats. J. Surg. Res. 2014, 187, 646–652. [Google Scholar] [CrossRef]
  79. Schumann, R.R. Malarial fever: Hemozoin is involved but Toll-free. Proc. Natl. Acad. Sci. USA 2007, 104, 1743–1744. [Google Scholar] [CrossRef] [PubMed]
  80. Chan, M.M.Y. Inhibition of tumor necrosis factor by curcumin, a phytochemical. Biochem. Pharmacol. 1995, 49, 1551–1556. [Google Scholar] [CrossRef]
  81. Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian solid gold. The molecular targets and therapeutic uses of curcumin in health and disease. AEMB 2007, 595, 1–75. [Google Scholar]
  82. Siebenlist, U.; Franzoso, G.; Brown, K. Structure, regulation and function of NF-kappaB. Annu. Rev. Cell Dev. Biol. 1994, 10, 405–455. [Google Scholar] [CrossRef]
  83. Shishodia, S.; Singh, T.; Chaturvedi, M.M. Modulation of transcription factors by curcumin. The molecular targets and therapeutic uses of curcumin in health and disease. AEMB 2007, 595, 127–148. [Google Scholar]
  84. Xu, Y.X.; Pindolia, K.R.; Janakiraman, N.; Noth, C.J.; Chapman, R.A.; Gautam, S.C. Curcumin, a compound with anti-inflammatory and anti-oxidant properties, down-regulates chemokine expression in bone marrow stromal cells. Exp. Hematol. 1997, 25, 413–422. [Google Scholar]
  85. Surh, Y.J. Anti-tumor promoting potential of selected spice ingredients with antioxidative and anti-inflammatory activities: A short review. Food Chem. Toxicol. 2002, 40, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
  86. Paulino, N.; Paulino, A.S.; Diniz, S.N.; de Mendonça, S.; Gonçalves, I.D.; Flores, F.F.; Santos, R.P.; Rodrigues, C.; Pardi, P.C.; Suarez, J.A.Q. Evaluation of the anti-inflammatory action of curcumin analog (DM1): Effect on iNOS and COX-2 gene expression and autophagy pathways. Bioorg. Med. Chem. 2016, 24, 1927–1935. [Google Scholar] [CrossRef] [PubMed]
  87. Perlmann, P.; Troye-Blomberg, M. Malaria blood-stage infection and its control by the immune system. Folia Biol. 2000, 46, 210–218. [Google Scholar]
  88. Jang, M.K.; Sohn, D.H.; Ryu, J.H. A curcuminoid and sesquiterpenes as inhibitors of macrophage TNF-α release from Curcuma zedoaria. Planta Med. 2001, 67, 550–552. [Google Scholar] [CrossRef] [PubMed]
  89. Matsuda, H.; Tewtrakul, S.; Morikawa, T.; Nakamura, A.; Yoshikawa, M. Anti-allergic principles from Thai zedoary: Structural requirements of curcuminoids for inhibition of degranulation and effect on the release of TNF-α and IL-4 in RBL-2H3 cells. Bioorg. Med. Chem. 2004, 12, 5891–5898. [Google Scholar] [CrossRef]
  90. Sarkar, F.H.; Li, Y. Cell signaling pathways altered by natural chemopreventive agents. Mutat. Res.-Fundam. Mol. Mech. Mutagen. 2004, 555, 53–64. [Google Scholar] [CrossRef] [PubMed]
  91. Singh, S.; Aggarwal, B.B. Activation of transcription factor NF-κB is suppressed by curcumin (diferuloylmethane). J. Biol. Chem. 1995, 270, 24995–25000. [Google Scholar] [CrossRef]
  92. Ali, A.H.; Sudi, S.; Basir, R.; Embi, N.; Sidek, H.M. The antimalarial effect of curcumin is mediated by the inhibition of glycogen synthase kinase-3 β. J. Med. Food. 2017, 20, 152–161. [Google Scholar] [CrossRef]
  93. Mimche, P.N.; Taramelli, D.; Vivas, L. The plant-based immunomodulator curcumin as a potential candidate for the development of an adjunctive therapy for cerebral malaria. Malar. J. 2011, 10, 510. [Google Scholar] [CrossRef]
  94. Kaiser, K.; Texier, A.; Ferrandiz, J.; Buguet, A.; Meiller, A.; Latour, C.; Peyron, F.; Cespuglio, R.; Picot, S. Recombinant human erythropoietin prevents the death of mice during cerebral malaria. J. Infect. Dis. 2006, 193, 987–995. [Google Scholar] [CrossRef]
  95. Cui, L.; Miao, J.; Cui, L. Cytotoxic effect of curcumin on malaria parasite Plasmodium falciparum: Inhibition of histone acetylation and generation of reactive oxygen species. Antimicrob. Agents Chemother. 2007, 51, 488–494. [Google Scholar] [CrossRef]
  96. Shehzad, A.; Qureshi, M.; Anwar, M.N.; Lee, Y.S. Multifunctional curcumin mediate multitherapeutic effects. J. Food Sci. 2017, 82, 2006–2015. [Google Scholar] [CrossRef] [PubMed]
  97. Banik, U.; Parasuraman, S.; Adhikary, A.K.; Othman, N.H. Curcumin: The spicy modulator of breast carcinogenesis. J. Exp. Clin. Cancer Res. 2017, 36, 98. [Google Scholar] [CrossRef] [PubMed]
  98. Hassan, F.U.; Rehman, M.S.U.; Khan, M.S.; Ali, M.A.; Javed, A.; Nawaz, A.; Yang, C. Curcumin as an alternative epigenetic modulator: Mechanism of action and potential effects. Front. Genet. 2019, 10, 514. [Google Scholar] [CrossRef] [PubMed]
  99. Bhaumik, S.; Anjum, R.; Rangaraj, N.; Pardhasaradhi, B.V.V.; Khar, A. Curcumin mediated apoptosis in AK-5 tumor cells involves the production of reactive oxygen intermediates. FEBS Lett. 1999, 456, 311–314. [Google Scholar] [CrossRef] [PubMed]
  100. Fujisawa, S.; Atsumi, T.; Ishihara, M.; Kadoma, Y. Cytotoxicity, ROS-generation activity and radical-scavenging activity of curcumin and related compounds. Anticancer Res. 2004, 24, 563–570. [Google Scholar] [PubMed]
  101. Alumasa, J.N.; Gorka, A.P.; Casabiance, L.B.; Comstock, E.; de Dios, A.C.; Roepe, P.D. The hydroxyl functionality and a rigid proximal N are required for forming a novel non-covalent quinine-heme complex. J. Inorg. Biochem. 2011, 105, 467. [Google Scholar] [CrossRef]
  102. Hunt, N.H.; Golenser, J.; Chan-Ling, T.; Parekh, S.; Rae, C. Immunopathogenesis of cerebral malaria. Int. J. Parasitol. 2006, 36, 569–582. [Google Scholar] [CrossRef]
  103. Schofield, L.; Grau, G.E. Immunological processes in malaria pathogenesis. Nat. Rev. Immunol. 2005, 5, 722–735. [Google Scholar] [CrossRef]
  104. Balaji, S.N.; Ahsan, M.J.; Jadav, S.S.; Trivedi, V. Molecular modelling, synthesis, and antimalarial potentials of curcumin analogues containing heterocyclic ring. Arab. J. Chem. 2015, 12, 2492–2500. [Google Scholar] [CrossRef]
  105. Parveen, A.; Chakraborty, A.; Konreddy, A.K.; Chakravarty, H.; Sharon, A.; Trivedi, V.; Bal, C. Skeletal hybridization and PfRIO-2 kinase modeling for synthesis of α-pyrone analogs as anti-malarial agent. Eur. J. Med. Chem. 2013, 70, 607–612. [Google Scholar] [CrossRef]
  106. Gui, J.S.; Jalil, J.; Jubri, Z.; Kamisah, Y. Parkia speciosa empty pod extract exerts anti-inflammatory properties by modulating NFκB and MAPK pathways in cardiomyocytes exposed to tumor necrosis factor-α. Cytotechnology 2019, 71, 79–89. [Google Scholar] [CrossRef] [PubMed]
  107. Singh, D.B.; Dwivedi, S. Structural insight into binding mode of inhibitor with SAHH of Plasmodium and human: Interaction of curcumin with anti-malarial drug targets. J. Chem. Biol. 2016, 9, 107–120. [Google Scholar] [CrossRef] [PubMed]
  108. Zahidah, A.F.; Faizah, O.; Nur Aqilah, K.; Taty Anna, K. Curcumin as an anti-arthritic agent in collagen-induced arthritic Sprague-Dawley rats. Sains Malays. 2012, 41, 591–595. [Google Scholar]
  109. Kevin, T.T.M.; Nur Idanis, A.S.; Anastasha, B.; Mohd Faris, M.R.; Faizah, O.; Taty Anna, K. Curcumin minimises histopathological and immunological progression in the ankle joints of collagen-induced arthritis rats. Med. Health 2020, 15, 26–36. [Google Scholar]
  110. Kamal, D.A.M.; Salamt, N.; Yusuf, A.N.M.; Kashim, M.I.A.M.; Mokhtar, M.H. Potential health benefits of curcumin on female reproductive disorders: A review. Nutrients 2021, 13, 3126. [Google Scholar] [CrossRef] [PubMed]
  111. Irfandi, R.; Ilham, M.; Erwing, R.; Arafah, M.; Rompegading, A.B.; Putri, S.E.; Sartika, S.D.; Fauziah, S.; Agustina, A.S.; Akbar, H.; et al. Review on curcumin compounds in turmeric plants for the treatment of COVID-19. Int. J. Des. Nat. Ecodyn. 2022, 17, 957–965. [Google Scholar] [CrossRef]
  112. Waknine-Grinberg, J.H.; McQuillan, J.A.; Hunt, N.; Ginsbur, J. Modulation of cerebral malaria by fasudil and other immune-modifying compounds. Exp. Parasitol. 2010, 125, 141–146. [Google Scholar] [CrossRef]
  113. Khairani, S.; Fauziah, N.; Wiraswati, H.L.; Panigoro, R.; Salleh, A.; Setyowati, E.Y.; Berbudi, A. Piperine enhances the antimalarial activity of curcumin in Plasmodium berghei ANKA-infected mice: A novel approach for malaria prophylaxis. Evid.-Based Complement. Altern. Med. 2022, 2022, 7897163. [Google Scholar] [CrossRef]
  114. Alkandahri, M.Y.; Berbudi, A.; Subarnas, A. Evaluation of experimental cerebral malaria of curcumin and kaempferol in Plasmodium berghei ANKA-infected mice. Pharmacogn. J. 2022, 14, 905–911. [Google Scholar] [CrossRef]
  115. Balasaheb, R.; Wanare, G.; Kawathekar, N.; Ranjan, R.; Kumar, N.; Sahal, D.; Singh, V. Dibenzylideneacetone analogues as novel Plasmodium falciparum inhibitors. Bioorganic Med. Chem. Lett. 2011, 21, 3034–3036. [Google Scholar]
  116. Munigunti, R.; Gathiaka, S.; Acevedo, O.; Sahu, R.; Tekwani, B.; Calderón, A.I. Determination of antiplasmodial activity and binding affinity of curcumin and demethoxycurcumin towards PfTrxR. Nat. Prod. Res. 2014, 28, 356–364. [Google Scholar] [CrossRef] [PubMed]
  117. Singh, D.B.; Gupta, M.K.; Singh, D.V.; Singh, S.K.; Misra, K. Docking and in silico ADMET studies of noraristeromycin, curcumin and its derivatives with Plasmodium falciparum SAH hydrolase: A molecular drug target against malaria. Interdiscip. Sci. Comput. Life Sci. 2013, 5, 1–12. [Google Scholar] [CrossRef] [PubMed]
  118. Mimche, P.N.; Thompson, E.; Taramelli, D.; Vivas, L. Curcumin enhances non-opsonic phagocytosis of Plasmodium falciparum through up-regulation of cd36 surface expression on monocytes/macrophages. J. Antimicrob. Chemother. 2012, 67, 1895–1904. [Google Scholar] [CrossRef] [PubMed]
  119. Montesino, N.L.; Kaiser, M.; Brun, R.; Schmidt, T.J. Search for antiprotozoal activity in herbal medicinal preparations; new natural leads against neglected tropical diseases. Molecules 2015, 20, 14118–14138. [Google Scholar] [CrossRef] [PubMed]
  120. Ullah, R.; Rehman, A.; Zafeer, M.F.; Rehman, L.; Khan, Y.A.; Khan, M.A.H.; Khan, S.N.; Khan, A.U.; Abidi, S.M.A. Anthelmintic potential of thymoquinone and curcumin on Fasciola gigantica. PLoS ONE 2017, 12, e0171267. [Google Scholar] [CrossRef]
  121. Busari, Z.A.; Dauda, K.A.; Morenikeji, O.A.; Afolayan, F.; Oyeyemi, O.T.; Meena, J.; Sahu, D.; Panda, A.K. Antiplasmodial activity and toxicological assessment of curcumin PLGA-encapsulated nanoparticles. Front. Pharmacol. 2017, 8, 622. [Google Scholar] [CrossRef]
  122. Memvanga, P.B.; Coco, R.; Préat, V. An oral malaria therapy: Curcumin-loaded lipid-based drug delivery systems combined with β-arteether. J. Control. Release 2013, 172, 904–913. [Google Scholar] [CrossRef]
  123. Olanlokun, J.O.; Abiodun, W.O.; Ebenezer, O.; Koorbanally, N.A.; Olorunsogo, O.O. Curcumin modulates multiple cell death, matrix metalloproteinase activation and cardiac protein release in susceptible and resistant Plasmodium berghei-infected mice. Biomed. Pharmacother. 2022, 146, 112454. [Google Scholar] [CrossRef]
Figure 1. The life cycle of the Plasmodium parasite, beginning from the infection of sporozoites released by the bite of a female Anopheles mosquito into the circulatory system of humans [1,4].
Figure 1. The life cycle of the Plasmodium parasite, beginning from the infection of sporozoites released by the bite of a female Anopheles mosquito into the circulatory system of humans [1,4].
Pharmaceuticals 16 00609 g001
Figure 2. Currently available drugs and biologically active compounds used in the research for treatment of malaria. [2,3,8,9].
Figure 2. Currently available drugs and biologically active compounds used in the research for treatment of malaria. [2,3,8,9].
Pharmaceuticals 16 00609 g002
Figure 3. The chemical structures of curcumin and two other curcuminoids naturally present in commercially available curcumin.
Figure 3. The chemical structures of curcumin and two other curcuminoids naturally present in commercially available curcumin.
Pharmaceuticals 16 00609 g003
Figure 4. Chemical structure of curcumin with its two methoxy phenolic groups.
Figure 4. Chemical structure of curcumin with its two methoxy phenolic groups.
Pharmaceuticals 16 00609 g004
Figure 5. The reaction scheme for the synthesis of di-O-acetyl and di-methoxycurcumin involving substitution of the phenolic OH group [37].
Figure 5. The reaction scheme for the synthesis of di-O-acetyl and di-methoxycurcumin involving substitution of the phenolic OH group [37].
Pharmaceuticals 16 00609 g005
Figure 6. The equilibrium between the (i) keto and (ii) enolate forms of curcumin due to the reactivity of the methylene α -hydrogen, which is susceptible to nucleophilic attack, leading to the low stability of curcumin [45,53,54].
Figure 6. The equilibrium between the (i) keto and (ii) enolate forms of curcumin due to the reactivity of the methylene α -hydrogen, which is susceptible to nucleophilic attack, leading to the low stability of curcumin [45,53,54].
Pharmaceuticals 16 00609 g006
Figure 7. Specific sites and functional groups of curcumin, which can be targeted for structural modifications.
Figure 7. Specific sites and functional groups of curcumin, which can be targeted for structural modifications.
Pharmaceuticals 16 00609 g007
Figure 8. Examples of curcumin derivatives with a wide range of therapeutic properties [9,38,50,55,56,57,58,59,60,61].
Figure 8. Examples of curcumin derivatives with a wide range of therapeutic properties [9,38,50,55,56,57,58,59,60,61].
Pharmaceuticals 16 00609 g008
Figure 9. Chemical structures of curcumin derivatives, which have been synthesized and studied for their anti-inflammatory and antimalarial activities [23,47].
Figure 9. Chemical structures of curcumin derivatives, which have been synthesized and studied for their anti-inflammatory and antimalarial activities [23,47].
Pharmaceuticals 16 00609 g009aPharmaceuticals 16 00609 g009b
Figure 10. Reaction scheme showing the synthesis of isoxazole and pyrazole derivatives of curcumin.
Figure 10. Reaction scheme showing the synthesis of isoxazole and pyrazole derivatives of curcumin.
Pharmaceuticals 16 00609 g010
Figure 11. The pyrazole derivative of curcumin has higher efficacy than the isoxazole derivative.
Figure 11. The pyrazole derivative of curcumin has higher efficacy than the isoxazole derivative.
Pharmaceuticals 16 00609 g011
Figure 12. Pyrazole derivatives of curcumin with a phenyl group attached to the nitrogen atom. The presence of different groups, either electron donating (EDG) or electron withdrawing (EWG), on the phenyl group varyingly influences the bioactivity of the compounds.
Figure 12. Pyrazole derivatives of curcumin with a phenyl group attached to the nitrogen atom. The presence of different groups, either electron donating (EDG) or electron withdrawing (EWG), on the phenyl group varyingly influences the bioactivity of the compounds.
Pharmaceuticals 16 00609 g012
Figure 13. Pyrazole derivative (6) with a nitro substituent at the phenyl ring, which contributes to its enhanced bioactivity.
Figure 13. Pyrazole derivative (6) with a nitro substituent at the phenyl ring, which contributes to its enhanced bioactivity.
Pharmaceuticals 16 00609 g013
Figure 14. The reaction scheme and the chemical structures of derivatives (14) and (15), as synthesized by Knoevenagel condensation [37,38,70].
Figure 14. The reaction scheme and the chemical structures of derivatives (14) and (15), as synthesized by Knoevenagel condensation [37,38,70].
Pharmaceuticals 16 00609 g014
Figure 15. Curcumin derivative whose active methylene group is substituted by Knoevenagel condensation.
Figure 15. Curcumin derivative whose active methylene group is substituted by Knoevenagel condensation.
Pharmaceuticals 16 00609 g015
Figure 16. Structure of curcumin derivative (11) with both EDG and EWG groups attached to the additional phenyl.
Figure 16. Structure of curcumin derivative (11) with both EDG and EWG groups attached to the additional phenyl.
Pharmaceuticals 16 00609 g016
Figure 17. Curcumin derivative (21) had the highest potency among the synthesized Knoevenagel condensate derivatives.
Figure 17. Curcumin derivative (21) had the highest potency among the synthesized Knoevenagel condensate derivatives.
Pharmaceuticals 16 00609 g017
Figure 18. Reaction scheme showing the synthesis of monocarbonyl curcumin derivatives (4347).
Figure 18. Reaction scheme showing the synthesis of monocarbonyl curcumin derivatives (4347).
Pharmaceuticals 16 00609 g018
Figure 19. Targeted structural modification sites of monocarbonyl curcumin derivatives.
Figure 19. Targeted structural modification sites of monocarbonyl curcumin derivatives.
Pharmaceuticals 16 00609 g019
Figure 20. Cellular level mechanism of action of malarial infection in the host cell involving the activation of NF- κ B [79].
Figure 20. Cellular level mechanism of action of malarial infection in the host cell involving the activation of NF- κ B [79].
Pharmaceuticals 16 00609 g020
Figure 21. The structural flexibility of curcumin allows for bond rotation around the α -carbon, bridging the two carbonyl groups.
Figure 21. The structural flexibility of curcumin allows for bond rotation around the α -carbon, bridging the two carbonyl groups.
Pharmaceuticals 16 00609 g021
Figure 22. Examples of potential parasite target proteins involved in the pathophysiology of malaria.
Figure 22. Examples of potential parasite target proteins involved in the pathophysiology of malaria.
Pharmaceuticals 16 00609 g022
Table 1. Physicochemical and pharmacokinetic properties of each compound shown in Figure 7, as generated by the Swiss ADME server [62].
Table 1. Physicochemical and pharmacokinetic properties of each compound shown in Figure 7, as generated by the Swiss ADME server [62].
CompoundPhysicochemical PropertiesLipophilicityWater SolubilityDrug-LikenessPharmacokinetics
Molecular Weight (g/mol)Heavy AtomsRotatable BondsH-Bond AcceptorsH-Bond DonorsLog PLog SLipinski’s Rule ViolationGI AbsorptionBBB Permeability
1368.38278623.27 3.94NoHighNo
2365.38276623.60 4.33NoHighNo
3366.41276533.47 4.33NoHighNo
4460.65339534.45 4.95NoHighYes
5478.64349634.53 5.12NoHighYes
6505.653610733.79 5.051HighNo
7529.54359534.82 6.161HighNo
8490.683510634.72 5.05NoHighNo
9456.49349623.61 5.83NoHighNo
10472.49359733.55 5.69NoHighNo
11502.513710834.34 5.771LowNo
12452.453312803.83 4.28NoHighNo
13396.432910603.59 4.37NoHighYes
14472.49359733.56 5.69NoHighNo
15472.49359733.37 5.69NoHighNo
16488.49369843.44 5.56NoLowNo
17504.48379953.63 5.421LowNo
18486.513610724.11 5.91NoHighNo
19486.513610723.95 5.91NoHighNo
20516.543811823.86 5.991HighNo
21474.48359723.76 5.99NoHighNo
22492.47369823.65 6.15NoHighNo
23470.51359623.62 6.13NoHighNo
24484.54369623.90 6.44NoHighNo
25484.54369623.97 6.44NoHighNo
26490.93359623.82 6.42NoHighNo
27525.38369623.48 7.021HighNo
28501.483710822.99 5.891LowNo
29499.553710624.22 6.07NoHighNo
30501.483710822.76 5.891LowNo
31498.573710623.89 6.69NoHighNo
32500.54379733.67 6.301LowNo
33498.57379623.61 6.74NoHighNo
34500.543711724.50 6.141HighNo
35498.57379623.60 6.74NoHighNo
36484.543610624.19 6.41NoHighNo
37488.50369723.44 6.29NoHighNo
38488.50369723.86 6.29NoHighNo
39481.50369723.36 5.78NoHighNo
40471.50359633.38 5.47NoHighNo
41546.4840111022.14 5.972LowNo
42502.583610624.66 6.351LowNo
43326.34246523.10 3.95NoHighYes
44266.29204322.17 3.93NoHighYes
45324.29246502.18 4.22NoHighNo
46294.34226303.46 4.39NoHighYes
47392.08204103.61 6.111HighYes
GI—gastrointestinal; BBB—blood–brain barrier.
Table 2. Data reported by Mishra et al. [38] showing the in vitro efficacy of curcumin and its derivatives against P. falciparum.
Table 2. Data reported by Mishra et al. [38] showing the in vitro efficacy of curcumin and its derivatives against P. falciparum.
CompoundP. falciparum In Vitro Analysis
Chloroquine-Sensitive
FCK2
Chloroquine-Resistant
MP-14
IC50 (μM)IC50 (μM)
13.254.21
28.447.92
30.480.45
48.489.10
52.422.10
60.870.89
74.654.80
822.6024.56
93.894.12
105.855.36
110.920.75
122.342.51
137.868.40
Table 3. In vitro and in silico assessment of antimalarial activity of curcumin derivatives by Dohutia et al. [37].
Table 3. In vitro and in silico assessment of antimalarial activity of curcumin derivatives by Dohutia et al. [37].
CompoundMW (g/mol)logPH-Bond DonorH-Bond AcceptorSolubility (mg/L)IC50
( μ M)
Free Binding Energy (kcal/mol)% Schizont
Inhibition
5 μg/mL50 μg/mL
(21)474.485.43261382.51-−6.7597.8100
(15)472.494.97371869.61-−5.8989.5100
(9)456.495.33261583.743.89−5.3580.1100
Curcumin368.383.29267.4753.25−5.2579.6100
(10)472.494.97371869.615.85−3.87--
Table 4. Antimalarial and anti-inflammatory activities of curcumin and its derivatives.
Table 4. Antimalarial and anti-inflammatory activities of curcumin and its derivatives.
ActivityIn Vitro/In Vivo/In Silico EvidenceReferences
Antiplasmodium PfATP6Curcumin (1) reduced P. falciparum viability, causing parasitic cell proliferation to decrease.
-
Reported IC50 value: 5 μM.
[69]
Curcumin (1) and its derivatives (9, 14, 15, 19, 21, 23, 27, and 28) showed 100% inhibition of P. falciparum growth upon a 50 μ g/mL dose of treatment.[37]
Molecular docking results validated binding of curcumin (1) and its derivatives to PfATP with favorable free binding energy.
-
Reported free binding energy:
Curcumin derivative (21): –6.75 kcal/mol
(higher than both artemisinin (–6.73 kcal/mol) and curcumin (–5.25 kcal/mol), hence, better interaction with the protein).
-
Hydrogen bonding with Lys1213 and Leu1044
[37]
In vitro study using CQR P. falciparum showed potent antimalarial activity of curcumin (1), with reported IC50 value of ~5 μ M.
In vivo treatment of P. berghei-infected mice with 100 mg/kg curcumin showed:
-
More than 80% inhibition of parasitic growth.
-
A 29% increase in survival rate.
[23,25]
Curcumin treatment on P. berghei-infected C57BI/6 mice delayed mice death by 10 days and prevented cerebral malaria.
Dose: 50 mg/kg, twice daily for 6 days.
[112]
Curcumin exhibited antimalarial activity in P. berghei-infected mice.
Dose: 300 mg/kg daily for 4 days (60.22% parasitemia inhibition).
Dose: 80 mg/kg daily for 4 days (60.21% chemosuppressive effect).
[113,114]
Antiplasmodium Pf3D7Curcumin (1) showed potential inhibition of parasite transmission at the trophozoite stage.
Curcumin derivative (monocarbonyl curcumin)
-
Reported IC50 values against CQS: 1.97 μM, CQR: 1.69 μM.
Curcumin derivative (13)
-
Reported IC50 value: 1.97 μM.
[115]
Antiplasmodium PfDXRIn silico and in vitro studies validated synergistic binding of curcumin (1) to PfDXR protein with fosmidomycin.
-
Presence of methoxy substituent on the phenyl groups facilitated parasite elimination:
 (43)—55%.
 (46)—57%.
[74]
Antiplasmodium
PGCN5 HAT
In vitro study suggested curcumin (1) as a potent inhibitor of p300/CBP (CREB-binding protein) as tested on four P. falciparum strains.
-
Reported IC50 values—3D7: 24.69 μM, D10: 22.93 μM, 7G8: 29.61 μM, Dd2: 27.45 μM.
[92]
Antiplasmodium
PfTrxR
In vitro study using CQS (D6 clone) and CQR (W2 clone) P. falciparum strains showed that curcumin (1) inhibited PfTrxR protein with an IC50 value of 2 μ M.[116]
Antiplasmodium
PfHGPRT
PfSAHH
In silico simulation using Molegro Virtual Docker (MVD) and admetSAR showed high binding energy of curcumin (1) to the protein.
-
Reported binding energy:
-
PfHGPRT: −175.97 kcal/mol.
-
PfSAHH: −138.30 kcal/mol.
[107,117]
Antimalaria
ROS
In vitro study showed that curcumin (1) induced intracellular ROS production related to PPARɣ/Nrf2 activation.
-
Reported IC50 value: 10 μM.
[95,118]
AntimalariaIn vitro study using NF54 intraerythrocytic-form P. falciparum strain reported highly potent antiparasitic activity of curcumin (1).
-
Reported IC50 value: 0.59 μM.
[119]
In vitro study using 3D7 clone strain of P. falciparum reported synergistic antimalarial effect of curcumin (1) with dihydroartemisinin and reduction in hemozoin formation upon several consecutive treatments of curcumin.
-
Reported IC50 value: 2.2 μg/mL.
[100]
In vitro study showed the effectiveness of curcumin–artemisinin combination therapy with additive interaction in killing P. falciparum.
In vivo study using P. berghei-infected mice showed 100% survival upon treatment.
Dose: 750 µg.
[119]
In vivo study on P. berghei ANKA-infected mice revealed treatment of curcumin (1) reduced parasitemia level and increased survival rate.
Dose: 50 mg/kg daily.
[120]
In vitro study shows reported IC50:
-
Curcumin Encapsulated to PLGA: 292.6 µg/mL.
-
Curcumin (1): 1000 µg/mL.
In vivo study on P. berghei-infected mice and murine RAW 264.7 macrophages using curcumin encapsulated in PLGA showed 56.8% parasite suppression (higher than free curcumin with 40.5% suppression).
Dose: 5 and 10 mg/kg.
[121]
Anti-inflammatory
COX-2
In vitro study using DPPH radical-scavenging assay showed anti-inflammatory activity of curcumin and its derivatives.
Reported IC50 value and % inhibition:
-
Curcumin (1)—IC50 value: 11.06 μM, 35.0% inhibition.
-
Derivative (2)—IC50 value: 10.71 μM, 58.1% inhibition.
-
Derivative (3)—IC50 value: 9.70 μM, 61.0% inhibition.
[65]
Molecular docking using FlexX program validated COX-2 as a target protein and showed binding of curcumin (1) and curcumin derivatives (2, 3).
Favorable interactions:
-
Hydrogen bonding interaction with Arg120.
-
van der Waals interactions with Val523, Val116, Ala516, and Try355.
[65]
Anti-inflammatory
NF- κ B
In vivo study on P. berghei ANKA-infected mice upon treatment of curcumin (1) showed inhibition of NF- κ B activation, which reduced expression of adhesion molecules and suppressed pro-inflammatory cytokines level.
Dose: 100 mg/kg daily for 4 days.
[122]
Anti-inflammatory
GSK3β
In vivo study on P. berghei NK65-infected rats upon treatment of curcumin (1) showed inhibition of host GSK3 β , leading to the phosphorylation of NF- κ B and regulation of pro- (decrease in serum TNF-α and IFN-γ levels) and anti-inflammatory (IL-10 and IL-4) cytokines.
Dose: 3, 10, and 30 mg/kg.
[88]
Anti-inflammatoryIn vivo study on P. berghei NK65- and ANKA-infected mice upon treatment of curcumin (1).
Reported activity:
-
Significant decrease in inflammatory cytokine levels including serum p53, TNF-α, CRP, and IL-6.
-
Inhibition of mPT pore opening, F0F1 ATPase activity and mLPO.
Dose: 50 mg/kg.
[123]
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

Jamil, S.N.H.; Ali, A.H.; Feroz, S.R.; Lam, S.D.; Agustar, H.K.; Mohd Abd Razak, M.R.; Latip, J. Curcumin and Its Derivatives as Potential Antimalarial and Anti-Inflammatory Agents: A Review on Structure–Activity Relationship and Mechanism of Action. Pharmaceuticals 2023, 16, 609. https://doi.org/10.3390/ph16040609

AMA Style

Jamil SNH, Ali AH, Feroz SR, Lam SD, Agustar HK, Mohd Abd Razak MR, Latip J. Curcumin and Its Derivatives as Potential Antimalarial and Anti-Inflammatory Agents: A Review on Structure–Activity Relationship and Mechanism of Action. Pharmaceuticals. 2023; 16(4):609. https://doi.org/10.3390/ph16040609

Chicago/Turabian Style

Jamil, Siti Nur Hidayah, Amatul Hamizah Ali, Shevin Rizal Feroz, Su Datt Lam, Hani Kartini Agustar, Mohd Ridzuan Mohd Abd Razak, and Jalifah Latip. 2023. "Curcumin and Its Derivatives as Potential Antimalarial and Anti-Inflammatory Agents: A Review on Structure–Activity Relationship and Mechanism of Action" Pharmaceuticals 16, no. 4: 609. https://doi.org/10.3390/ph16040609

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