Next Article in Journal
Extracellular Vesicles and Intercellular Communication: Challenges for In Vivo Molecular Imaging and Tracking
Next Article in Special Issue
Efficacy of FRO on Acne Vulgaris Pathogenesis
Previous Article in Journal
Development of Miconazole-Loaded Microemulsions for Enhanced Topical Delivery and Non-Destructive Analysis by Near-Infrared Spectroscopy
Previous Article in Special Issue
Pharmacotherapeutic Potential of Aloe secundiflora against Colorectal Cancer Growth and Proliferation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant Extracts as a Source of Natural Products with Potential Antimalarial Effects: An Update from 2018 to 2022

by
Giovane de Jesus Gomes Ribeiro
1,†,
Sun Liu Rei Yan
1,†,
Giuseppe Palmisano
2 and
Carsten Wrenger
1,*
1
Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
2
GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2023, 15(6), 1638; https://doi.org/10.3390/pharmaceutics15061638
Submission received: 7 March 2023 / Revised: 3 May 2023 / Accepted: 9 May 2023 / Published: 1 June 2023
(This article belongs to the Special Issue Pharmaceutical Applications of Plant Extracts)

Abstract

:
Malaria kills more than 500,000 people yearly, mainly affecting Africa and Southeast Asia. The disease is caused by the protozoan parasite from the genus Plasmodium, with Plasmodium vivax and Plasmodium falciparum being the main species that cause the disease in humans. Although substantial progress has been observed in malaria research in the last years, the threat of the spread of Plasmodium parasites persists. Artemisinin-resistant strains of this parasite have been reported mainly in Southeast Asia, highlighting the urgent need to develop more effective and safe antimalarial drugs. In this context, natural sources, mainly from flora, remain underexplored antimalarial spaces. The present mini-review explores this space focusing on plant extracts and some of their isolated natural products with at least in vitro antiplasmodial effects reported in the literature comprising the last five years (2018–2022).

1. Introduction

Malaria is still a worldwide public health problem, with an estimated 247 million cases and 619,000 deaths globally in 2022, principally affecting children in the African continent. Malaria primarily affects the intertropical region, where appropriate climatic and geographical conditions are suitable for the spread of the female Anopheles mosquito, the vector of the disease. The most prevalent species are Plasmodium falciparum and Plasmodium vivax, and the most severe form of the disease is caused by P. falciparum [1]. The rise of antimalarial resistance, mainly in Southeast Asia, due to the indiscriminate use of antimalarials and incomplete malaria drug treatment demonstrates the urgent need for the development of better and more efficacious drugs against the disease [2]. Although malaria prevention has shown considerable advances [3], the lack of an effective vaccine against the disease continues to increase the number of deaths in the African continent. In 2022, a ground-breaking malaria vaccine was prequalified by the WHO for the prevention of P. falciparum infections. This vaccine, named RTS, S/AS01 (Mosquirix) and developed in cooperation with the private pharmaceutical company GSK, was recommended by the WHO for the immunization of children from five months of age living in moderate- to high P. falciparum transmission regions in Africa. However, the effectiveness of this vaccine is not enough to completely halt the spread of severe malaria cases worldwide [4].
Despite the substantial public and private financial provision for malaria prevention resources and considerable scientific progress in the cellular, molecular, and clinical areas, chemotherapy remains a key tool for controlling the disease [5]. Malaria treatment has become increasingly complex, owing to the spread of artemisinin-resistant strains of Plasmodium. Recently, P. falciparum resistance to artemisinin derivatives has been detected in Southeast Asia [6,7]. As for P. vivax, the most important species on the African continent and in the Americas, there is a record of resistance to chloroquine and sulfadoxine-pyrimethamine in vivo and in vitro that has been increasingly documented [8,9].
Therefore, the current global malaria prevalence demonstrates the urgency for newer and more effective drugs against the disease [10]. Over the past millennium, different civilizations have used plants and their extracts for medicinal purposes. For example, the discovery of one of the most potent available antimalarials, artemisinin [11], was possible because of the almost 2000-year-old antimalarial recipe empirically used and written by ancient Chinese for traditional Chinese medicine. This demonstrates the importance of natural products and their derivatives for treating harmful human diseases.
Natural products have high structural diversity, biochemical specificity, and other molecular properties, making them promising lead compounds for developing new drugs [12].
Artemisinin was discovered in plant extracts (Artemisia annua); similarly, other antimalarials in therapeutics, such as quinine and quinidine, were first detected in the bark of the cinchona tree [13] more than 350 years ago in South America. The clear indication of promising antimalarials originated from plant extracts strongly suggests that new chemical entities may emerge from these valuable natural sources.

2. Malaria Scenario Worldwide: A Short Update

Malaria directly impacts communities’ health and economic development in tropical and subtropical regions. The World Health Organization (WHO) has estimated 247 million new cases in 2021. In 2021, 96% of malaria cases and deaths worldwide occurred in 29 of the 84 countries endemic to malaria (including the territory of French Guiana) [14]. Almost half of all cases came from four countries: Nigeria (26.6%), the Democratic Republic of the Congo (12.3%), Uganda (5.1%), and Mozambique (4.1%). Additionally, four countries accounted for just over half of all malaria deaths worldwide: Nigeria (31.3%), the Democratic Republic of the Congo (12.6%), the United Republic of Tanzania (4.1%), and Nigeria (3.9%). The percentage of total deaths from malaria among children under five years of age has decreased over the last 20 years, from 87.3% in 2000 to 76.8% in 2015; however, it has remained unchanged since then [15].
In 2021, the Southeast Asia region had nine countries where malaria is endemic, accounting for 5.4 million cases and contributing 2% of statistical malaria cases worldwide. India accounted for approximately 79% of all malaria cases worldwide in 2021. Approximately 40% of all cases in this region were caused by P. vivax. In the last 20 years, malaria cases have decreased by 76%, a reduction of about 17.4 million cases in 2021 [3]. An important recent achievement in the Asian continent was the inclusion of China as a certified malaria-free country.
In Latin America, Brazil, Colombia, and Venezuela are responsible for 79% of all malaria cases in the region. Most cases in this region are caused by P. vivax (71.5% in 2021).
The transmission of malaria in Brazil is restricted to the Legal Amazon, a territory that covers nine states: Acre, Amapá, Amazonas, Maranhão, Mato Grosso, Pará, Rondônia, Roraima, and Tocantins. This region accounts for approximately 99.7% of all malaria cases in the country. Two main species of Plasmodium are responsible for human malaria cases in Brazil: P. vivax and P. falciparum. Most reported cases were diagnosed as P. vivax infections, representing 83.0% (114,449) of registered cases, followed by P. falciparum infections, which accounted for 17.0% (23,408) of the cases [3].
As of 2020, epidemiological data indicate an annual decline in malaria cases, with 139,211 cases registered in 2021. The Legal Amazon is a territory where several indigenous communities live, including one of the largest indigenous groups, the Yanomami, who had approximately 11,530 cases of malaria in the last year, representing 9.93% of all malaria cases in Brazil [16]. However, there is a need for new interventions and improvements in disease surveillance and control.
There are also countries that achieved the WHO certification of malaria-free countries, including: Argentina (2019), El Salvador (2021), and Paraguay (2018), with zero cases of malaria for three consecutive years [17].

3. Methodology for Literature Selection and Previous Literature

For this mini-review, a database search was performed with Digital Science’s Dimensions platform (Dimensions AI), which also covers the PubMed database. The combination of keywords “Plasmodium” and “plant extract” in the title or abstract was used in the searches. The period covered is from 2018 to 2022. The search was initially conducted on 26 January 2023, but a few articles published after this date were also added due to the match of the keyword combination. The criteria for selecting the articles were primarily based on experimental data with a plant extract antiplasmodial activity less than 10 μg/mL. The articles were divided into two groups: those reporting natural products (divided into ethnobotanical studies and plant extracts) and those covering studies of isolated bioactive substances and possible pharmacological targets.
We have also analyzed recent reviews that referred to the use of medicinal plants as antimalarials [18,19,20,21,22,23]. They provide a thorough overview of the scientific literature with respect to the antimalarial effects of traditionally used medicinal plants, some of them mainly covering plants found in African countries, such as Ethiopia [20,22,23]. Another example is the review from Noronha and colleagues [21], which provides a broad revision of the literature concerning plants having antimalarial and antipyretic activity and the practical challenges of using these medicinal plants. In another review, Pan and collaborators [18] provide a comprehensive review covering more than 2000 plant extracts, including marine plants, and the discovery of 175 antimalarials from plants in the covered period of 2001–2017. The present mini-review differs from the cited previous literature, since it aims to provide an update on plant extracts and its most promising isolated compounds tested against Plasmodium falciparum, without restrictions to specific endemic plant species, and covering the last five years of scientific literature, as mentioned above.

4. Omics-Based Strategies for Targeted Isolation of Natural Products and Discovery of New Potential Drugs

Natural products play a key role in discovering new chemical entities [24], and plant extracts generally contain diverse chemical mixtures with different metabolites. Isolation of each chemical entity from these mixtures requires the use of combined analytical techniques, since the bioactive compounds are found in low concentrations and can also work in a synergistic mode. One challenging problem in the field of natural products is the identification of the mechanism of action (MOA) for small molecules. Target identification is crucial for compound optimization and further steps in the drug discovery process. However, it is also known that many drugs interact with different protein targets [25], which require broader target identification methods. In this sense, chemical proteomics may provide insights into the mechanism of action for small molecules. In chemical proteomics, the combination of chemistry, cellular biology, and mass spectrometry allows the generation of large data sets, with the possibility of understanding potential off-targets and how to generate a better lead compound. A crucial step in a chemical proteomics experiment is the probe design and synthesis, which depend on different probes, comprehensively revised by Chen and colleagues [25]. In chemical proteomics approaches, the natural products should be either chemically modified with an enrichment moiety (activity-based protein profiling) [26,27], linked to a resin, or conjugated with photoactivatable crosslinkers [28]. These approaches have been combined with quantitative label-free or label-based proteomics strategies to identify protein targets in complex biological systems. One drawback resides in the knowledge of structure–activity relationships, since chemical modifications can induce activity loss and binding to the wrong target.
The deconvolution of a protein target in an unbiased way has also been achieved using other approaches that explore protein stability upon small molecule binding. Protein stability can be challenged by raising the temperature, which will induce denaturation, aggregation, and precipitation. Proteins binding to a specific small molecule will respond differently to temperature changes. Thermal Proteome Profiling (TPP), cellular thermal shift assay coupled with mass spectrometry (CETSA®) [29], and Proteome Integral Solubility Alteration (PISA) [30] are based on thermal shifts. These assays can be performed in vitro, intact cell lines or cell lysates, and in vivo, in tissues and biofluids. MS-CETSA was applied to identify P. falciparum purine nucleoside phosphorylase as the target of quinine [31]. Another assay, termed drug affinity responsive target stability (DARTS), measures the resistance of a protein to hydrolysis by a broad specificity protease in the presence of a known small molecule [32]. The same principle was exploited in the limited proteolysis/LiP-small molecule mapping (LiP-SMap) [33]. Protein stability upon interaction with a small protein was also probed using the rate of methionine oxidation upon incubation with a denaturing agent. This assay was termed stability of proteins from rates of oxidation (SPROX) [34]. Recently, the resistance of a protein binding to a small molecule was probed using organic solvents. In this assay, termed solvent proteome profiling (SPP), the association of proteins with small molecule makes them more or less susceptible to solvent-induced denaturation [35]. The small molecule does not need to be modified in these assays, and its target(s) can be identified using a purified protein or a complex proteome. One drawback of these strategies is the solubility of membrane proteins. Indeed, the need to work with soluble proteins in their native state limits their applicability to multipass transmembrane proteins. Combining mass spectrometry-based proteomics strategies with these assays allows the identification of unknown targets.
Metabolomics has evolved to allow the analysis of thousands of metabolites from natural extracts [12], and it has been used to characterize metabolites of less than 1.5 kDa, such as amino acids and lipids, based mainly on nuclear magnetic resonance (NMR) and mass spectrometry (MS) data. It has already been used for the identification of drug targets, as well: early metabolomics studies permitted, for example, the development of FDA-approved drugs enasidenib (Idhifa®) and ivosidenib (Tibsovo®) for the treatment of acute myeloid leukemia [36,37,38,39].
Moreover, the application of metabolomics in the screening of natural product extracts via hyphenated techniques allows detailed structural characterization [40]. It also permits implementing databases that can be used to discover new bioactive natural products [41].
Recent developments in analytical chemistry include dereplication and high content screening (HCS). Dereplication refers to a process for the fast identification of known secondary metabolites in natural products, in which MS-based methods are key to the success of that process [42,43,44], and HCS methods are those that combine automated microscopy with living cell-based assays [45,46,47,48], which allows, e.g., the screening of plant extract libraries and subsequent selection of the best hits based on the phenotypic screening results. However, the isolation of chemical compounds from crude natural extracts remains a major challenge in the field [42]. The extraction method determines the chemical compound classes present in the purified fractions. Based on the polarity of the molecules present in the extract, using different organic solvents can selectively isolate a range of different compounds, directly affecting phytochemical isolation and yield. The combination of solvents would be a strategy to improve the extraction of compounds with different polarities [49], as illustrated in Figure 1. After identifying a crude extract with promising pharmacological activity, the extract is fractionated and guided by bioactivity until pure bioactive compounds are isolated [50].
Other limitations of crude extraction steps include the presence of known natural product molecules, chemical compounds without drug-like characteristics, and insufficient amounts of natural products for characterization [47]. These technical challenges can be addressed by the development of new methods using dereplication, extraction, and prefractionation of extracts [51]. Another approach is the use of metabolomics and genomics, in which data generated by molecular networks are analyzed and common characteristics of the natural extract composition are determined. The low yield in the extraction process and the structural elucidation of the chemical compounds are other aspects to be addressed in the field [47]. Bioinformatics tools and genomic engineering may also be helpful. Tandem mass spectrometry (MS/MS) libraries allow fast, reproducible, and accurate analysis of thousands of MS/MS spectra. Because there is a need for reliable information on MS/MS, different databases, including NIST, MassBank, MoNA European MassBank, LipidBlast, Wiley MSforID, and METLIN, are available for this purpose. In addition, the web platform Global Natural Product Social Molecular Networking allows the generation of a molecular network in which molecules are grouped based on their fragmentation pattern of similarity and similarity in the identification of metabolites [52].

5. Novel Antimalarials Derived from Plant Extracts and Isolated Natural Products

Antiplasmodial natural products are organized into families of promising extracts, and isolated compounds are organized in classes, such as alkaloids, terpenes, and polyphenols, with subclasses when applicable. The in vitro antiplasmodial activities described in this review were determined using various strains of P. falciparum with different drug sensitivities. The chloroquine-sensitive strains were 3D7, NF54, D6, HB3, D10, and TM4/8.2. Other strains used were the chloroquine-resistant strains Dd2, FcB1, and Pf INDO, as well as the multiresistant strains K1, W2, FCR3, and K1CB1. Different assays were used to determine in vitro antiplasmodial activity of the compounds. The most cited methods are the radioactive hypoxanthine incorporation assay, the colorimetric enzyme-linked immunosorbent assay (ELISA) that measures P. falciparum lactate protein dehydrogenase (pLDH), DNA-based fluorometric method using Pico Green Assay (SYBR), Malstat reagent, and microscopy. Regardless of the in vitro assay method, in this review, the inhibitory concentration range and extracts with half maximal inhibitory concentrations (IC50) ≤ 10 µg/mL were considered active [53]. Only isolated compounds with IC50 ≤ 10.0 µM were considered for further studies [54,55,56]. Based on these criteria, from the selected literature, a total of twenty isolated natural products derived from three different plant species were found to inhibit Plasmodium parasite proliferation in vitro (Table 1 and Figure 2).
Phytochemical investigation of aqueous and ethanolic leaf extracts of Azadirachta indica (Meliaceae) showed inhibition of P. falciparum with IC50 values of 7.4 µg/mL and 8.6 µg/mL, respectively. A study on the in vitro antimalarial activity of extracts from indigenous plant species in Kebbi State justifies the traditional use of the plant in malaria treatment, and further research may help identify and characterize the active principles of these plants [57].
The Homalolepis suffruticosa of the Simaroubaceae family is known for its popular use in treating malaria. Homalolepis Turcz. is a neotropical genus named Quinquina de Cayenne and represents one of the many drugs called ‘falsa quina’ (fake quine), which is used to treat malaria in South and Central America. In a bioassay-guided assay [58], the methanol extract of the roots of H. suffruticosa showed high in vitro antiplasmodial activity with an IC50 of 1.88 μg/mL in moderate cytotoxic HepG2 (CC50 = 41.93 μg/mL) and an SI = 22.30.
Goniothalamus lanceolatus (Annonaceae), an endemic plant from Borneo, is traditionally used in Malaysia to alleviate fever, as part of antimalarial treatment by folk medicine. Upon screening, methanol extract from roots exhibited potent antiplasmodial activity against both P. falciparum 3D7 (IC50 = 2.7 μg/mL, SI = 140) and K1 strains (IC50 = 1.7 μg/mL, SI = 236) [59]. The Annonaceae family is the largest family in Magnoliales order, comprising more than 100 genera and about 2400 species [76]. The large number of genus/species, with some of them reported in the literature as having antimalarial effects, such as those mentioned above for Goniothalamus lanceolatus, may also suggest further thorough investigation of potential antimalarials in that family [77].
According to a study by Dwivedi and collaborators [60], the plant Vitex negundo (Lamiaceae) is traditionally used for the treatment of malaria by indigenous tribes in certain countries, such as India and Malaysia. In vitro studies have reported the antimalarial use of the plant in traditional medicinal systems. The active extract of V. negundo leaf exhibited antimalarial activity with IC50 values of 7.21 μg/mL and 7.43 μg/mL against strains 3D7 and K1, respectively.
Petasites japonicus, an herb of the family Asteraceae, has been used to treat several human diseases. In Asian countries, the traditional use of P. japonicus is based on its many recognized pharmacological activities, including analgesic, anti-inflammatory, bactericidal, and antimalarial properties. A phytochemical study showed that the ethanolic extract from P. japonicus leaves significantly inhibited the parasite with IC50 values of 8.48 μg/mL and 7.83 μg/mL against strains 3D7 (CQ-sensitive) and Dd2, respectively [61]. The antiplasmodial activity of P. japonicus was also tested in vivo with P. berghei ANKA infected mice. The ethanolic extract of P. japonicus in a dose of 200 mg/kg lowered the parasitemia after four days of infection with a decrease percentage in the range from 65.21% to 97.14% [61].
In another study, the medicinal plant Senna occidentalis was evaluated for its phytochemical and activity against P. falciparum 3D7. The methanol extracts of the leaves and seeds of S. occidentalis (L.) (Fabaceae) (Leguminosae) showed significant activity against P. falciparum 3D7 with IC50 values of 12.19 µg/mL and 6.82 µg/mL, respectively [62]. In Kenya, the roots of the plant S. occidentalis are traditionally used for malaria treatment by the Luhya, Digo, and Duruma communities: the roots of the plant are decocted and taken orally three times a day over the course of three to four days. The methanolic extracts of S. occidentalis show antiplasmodial activity with a calculated IC50 of 1.76 μg/mL against 3D7 strain. No cytotoxicity was observed against a Vero cell line (CC50 247, IS of 140). The methanolic extract was also tested in vivo (P. berghei ANKA), showing an antiplasmodial effect of 200 mg/kg and effective dose (DE50) of 34.13 ± 5.14 mg/kg [78].
Plants of the genus Nauclea (Rubiaceae) are frequently used as folk medicines for the treatment of various diseases. Antimalarial activity of the hexane (IC50 = 1.93 μg/mL) and methanol (IC50 = 3.91 μg/ mL) fractions of Nauclea orientalis leaves against 3D7 P. falciparum was observed [63].
According to a previous investigation [64], the aqueous extracts of the leaves of the antimalarial Alchornea cordifolia (Euphorbiaceae) exhibited moderate activities against NF54 (chloroquine-sensitive parasite; IC50 of 5.8 μg/mL), CamWT_C580Y (artemisinin-sensitive parasite; IC50 17.8 μg/mL), and IPC 4912 (artemisinin-resistant parasite; IC50 15.8 μg/mL). The results of this study support the traditional use of the species A. cordifolia in Ghana as a folk remedy for treating malaria.
The Helianthus annuus (Asteraceae) plant is traditionally used as a medicinal remedy for several diseases, including malaria, in Oro-oro Ombo, Malang, East Java, and Indonesia. The ethanol extract of the roots and leaves of the species H. annuus exhibited antimalarial activity, with IC50 values of 2.3 and 4.3 μg/mL, respectively, against P. falciparum 3D7 [65]. The in vivo antiplasmodial effects of the ethanolic extract from H. annuus were also evaluated: 100 mg/kg with DE50 of 63.6 ± 8.0 mg/kg and 59.3 ± 13.2 mg/kg [65].
In Côte d’Ivoire, several traditional pharmacopeia plants, such as Pericopsis laxiflora and Harungana madagascariensis, have been used by various traditional practitioners for malaria treatment. The aqueous extract of the bark of H. madagascariensis (Hipperiacaceae) showed antiplasmodial activity, with an IC50 value of 6.16 µg/mL against P. falciparum NF54 [66]. The methanolic extracts of P. laxiflora (Euphorbiaceae) showed the best antiplasmodial activity, with an IC50 value of 7.44 µg/ mL against K1 [66].
Pleiocarpa, which belongs to the Apocynaceae family, is native to tropical Africa and is distributed from Senegal to Tanzania and Zimbabwe. Medicinal plants of this genus are well known for their use in the traditional treatment of pain, fever, and malaria. A study [67] investigated the dichloromethane/methanol (1:1) extract of Pleiocarpa bicarpellata roots, which showed strong antiplasmodial activity (IC50 = 3.5 µg/mL) against P. falciparum NF54. Negative cytotoxicity against L6 cells was observed.
Another recent study [58] on the Psychotria plant, which is native to Amazonia and Atlantic Forest biomes in Brazil, evaluated the antiplasmodial effect of the acid–base extracts of Psychotria apoda and P. colorata (Rubiaceae), which showed 90% inhibition of the parasite P. falciparum W2.
Andrographis paniculata is a tropical medicinal plant belonging to the Acanthaceae family. It is traditionally used for medicinal purposes in Hong Kong, Philippines, Taiwan, China, India, Malaysia, Indonesia, and many South Asian countries. The chloroform extract of whole plants of Andrographis paniculata exhibited antimalarial activity with IC50 values of 6.36 μg/mL and 5.24 μg/mL against strains 3D7 and K1, respectively, with no evidence of significant cytotoxicity against a mammalian cell line (CC50 > 100 μg/mL) [69].
Chaniad and collaborators [70] investigated the plant Globba malaccensis (Zingiberaceae), which is native to Nakhon Si Thammarat Province, Thailand. The ethanolic extract of G. malaccensis rhizomes showed promising antimalarial activity against P. falciparum K1 (IC50 = 1.50 µg/mL), with less toxicity to Vero cells (CC50 > 80 µg/mL). The ethanolic extract of this plant exhibited a considerable reduction in P. berghei parasitemia (murine model). A maximum suppressive effect of this extract (60.53%) was observed with the highest administered dose (600 mg/kg).
In Indonesia, the plant Sanchus arvensis L. (Asteraceae) is a traditional medicinal plant employed for malaria treatment. The ethyl acetate fraction obtained from leaves of S. arvensis showed in vitro antiplasmodial activity, with an IC50 of 2.92 μg/mL (3D7 strain), and it did not show cytotoxicity against human hepatic cells (Huh7it-1), with an SI of 97.03 [71]. The fraction was also tested in vivo (mice infected with P. berghei ANKA), showing an antiplasmodial activity below 100 mg/kg/day and ED50 46.31 ± 9.36 mg/kg. Chloroquine and chloroquine diphosphate were the positive controls for the in vitro studies, and DMSO served as the negative control.
The plant Terminalia arjuna (Roxb. ex DC.) (Wight and Arn.), known as “arjuna” or “Sa-mor-tes” in Thailand, has been traditionally used for various human diseases, given its antibacterial, antitumoral, and antiallergic effects. Against P. falciparum (strain K1), in vitro results showed an IC50 of 4.05 μg/mL [72]. In Vero cells, the CC50 was higher than 200 μg/mL, an indication of low cytotoxicity. For the in vitro study, the authors employed artesunate as a positive control and DMSO as a negative control. The in vivo study with aqueous extract of the T. arjuana fruits demonstrated a suppression of 67.95% of Plasmodium parasites at a dose of 600 mg/kg (P. berghei ANKA, murine model) [72]. Another plant species from the same genus, Terminalia bentzoe (L.) L.f. (Combretaceae) [73], was tested against P. falciparum (Dd2 strain), with an IC50 of 6.7 μg/mL and low cytotoxicity in Vero cells and CC50 207.03 μg/mL.
Another plant with potential antimalarial properties is Bridelia atroviridis Müll. Arg., a bush from Euphorbiaceae family that is found in equatorial Africa. This plant has been used in traditional medicine to treat various diseases, such as diabetes, gonorrhea, and fungal infections. Djouwoung and colleagues [74] have shown the antiplasmodial activity of hydroethanolic bark extract of Bridelia atroviridis Müll. Arg., exhibiting an in vitro antiplasmodial activity, with an IC50 of 8.08 μg/mL against the Dd2 strain. It also presented low cytotoxicity (CC50 > 100 μg/mL). The in vivo tests were performed in P. berghei NK 65 infected rats, resulting in a calculated ED50 of 89 mg/kg.
In Nigeria, the plant species Annickia affinis (Exell) (Versteegh and Sosef) from Annonaceae family is traditionally used for the treatment of fever and malaria. Decoction with water for this plant resulted in IC50 1.49 μg/mL (3D7 strain), while the CC50 was above 200 μg/mL [75]. In the in vivo test of infected mice with P. berghei ANKA, the A. affinis methanol extract at an oral dose of 100 mg/kg suppressed parasite growth by 32.46%. It was also investigated whether the plant has synergism with other plants as well, such as secondary metabolites from the mixture of plants named hepta-herbal Agbo-iba [75].
In another recent study, Ledoux and colleagues [79] investigated the potential antimalarial effects of the leaves of Casearia coriacea Vent. (Salicaceae), a plant endemic to the Mascarene Islands. Clerodane diterpenes were isolated from dichloromethane fraction and tested against the Plasmodium parasite: caseamembrin T, corymbulosine I, and isocaseamembrin E, with IC50 values of 0.25 µg/mL, 0.40 µg/mL, and 0.51 µg/mL, respectively. Cytotoxicity activity was considered slightly selective for pancreatic carcinoma (PANC-1). Structural determination was performed using NMR and mass spectrometry. This article is the first report of the isolation of these compounds from C. coriacea and their antiparasitic potentials. Due to the possible action of compound corymbulosine in apoptotic death, the compound could have its studies extended to cancer, as well. An interesting approach in this article is the fact that the authors have employed the zebrafish model as in vivo cytotoxicity assay rather than using mice model. Yet, the adjustment of the zebrafish model to test phytochemical compounds is also an open field.

6. Alkaloids: Potential Antimalarial Activity

Bioassay-guided fractionation of Hypoestes forskaolii (Vahl) R.Br. produced a phenanthroquinolizidine alkaloid, the isolated compound 15β-hydroxycryptopleurine-N-oxide (17), which has antiplasmodial activity. The chloroquine-resistant (K1) and chloroquine-sensitive (FCR3) P. falciparum strains were very sensitive to the antimalarial activity of 15β-hydroxycryptopleurine-N oxide, with IC50 values of 6.11 and 5.13 nM, respectively, while using chloroquine as the positive control [80]. A total of 15 mg was obtained from the plant, and a combination of techniques were used to purify the compound: silica gel column chromatography, high performance liquid chromatography (HPLC), and NMR (H, C, and two-dimensional).
From Cissampelos pareira (Linn) (Menispermaceae), a plant traditionally used to treat various diseases in India, a root ethyl acetate fraction with the most promising compound hayatinine (12), a bisbenzylisoquinoline alkaloid, was isolated (a total of 23 mg), having IC50 values of 0.41 μM (INDO) and 0.509 μM (3D7) [81]. Chloroquine was used as the positive control. Silica gel column chromatography, HPLC, NMR (H, C, and two-dimensional), and liquid chromatography coupled to mass spectrometry (LC/MS) were utilized for this alkaloid.
The phytochemical investigation of Aconitum heterophyllum (Wall. ex Royle) (Ranunculaceae) roots led to the identification of 2-O-cinnamoyl hetisine, a diterpenoid alkaloid in the chloroform fraction of the plant [82]. Antiplasmodial in vitro studies indicated an IC50 of 1.92 μM (INDO) and an IC50 10.8 μM (3D7). The yield of this compound was very low: from 2.6 g of chloroform fraction, only 16 mg of this hetisine were obtained.
The tazopsine alkaloid (compound 10 in Figure 2), isolated from Strychnopsis thouarsii Baill. (Menispermaceae), an endemic Malagasy plant, showed antiplasmodial activity and an of IC50, 7.88 µM against the NF54 strain, as well as a value of 4.07 µM against the 3D7 strain [83]. However, in P. yoelii infected mice, it was considered non-prophylactic, with 70% protection at a 100 mg/kg dose.

7. Terpenoids: Potential Antimalarial Activity

Sesquiterpene lactones have emerged as good starting points in the search for transmission-blocking drugs, with several studies demonstrating their potential. The most widely investigated member of this class of compounds is artemisinin, along with its derivatives. Greve and collaborators reported [84] that a triterpene derivative, compound rel-(16S,20S)-dihydroxydammar-24-en-3-one (16), had the most promising antimalarial activity. For this triterpene derivative, silica gel column chromatography, HPLC, NMR (H, C, and two-dimensional), and LC/MS were used.
The dichloromethane extract obtained from Commiphora myrrha Engl. (Burseraceae), the oleo-gum resin, showed promising in vitro activity against P. falciparum NF54, with an IC50 value of 1 µg/mL. Bioactivity-guided fractionation led to the isolation and characterization of 18 sesquiterpenoids. The sesquiterpene furanodienone (11) and the triterpene rel-(16S,20S)-dihydroxydammar-24-en-3-one (16) were the most active compounds found in this study, with IC50 values of 7.4 and 2.8 µM, respectively, with chloroquine as the positive control [84]. The techniques employed to isolate the compounds were silica gel column chromatography, HPLC, flash chromatography, NMR (H, C, and two-dimensional), and LC/MS.
Phytochemical investigation of the n-buthanolic extract of the roots of Terminalia albida (Sc. Elliot) (Combretaceae) by Balde and collaborators [85] led to the isolation and identification of ten oleanane triterpenoids, of which albidic acid B (18), albidic acid C (19), and albidienic acid (20) showed moderate antiplasmodial activity, with IC50 values ranging in the interval between 6 μM and 7 μM against K1 strain, also with chloroquine as the positive control. The isolation of the compounds was possible by employing the techniques of silica gel column chromatography, HPLC, NMR (H, C, and two-dimensional), and LC/MS.
The ethanolic extract of Cleistochlamys kirkii (Benth.) (Oliv.) (Annonaceae) root bark showed a 72% inhibition against chloroquine-sensitive 3D7-strain malarial parasite P. falciparum. The monotypic genus Cleistochlamys has a geographically restricted distribution in Southern Africa, and traditional uses of the plant comprise treatment of tuberculosis and rheumatism [86]. The isolated metabolites dichamanetin (4), (E)-acetylmelodorinol (5), and cleistenolide (6) showed IC50 values of 9.3, 7.6, and 15.2 µM, respectively, against P. falciparum 3D7, with chloroquine and artesunate as positive controls. Both the extract and the isolated compounds exhibited cytotoxicity against the negative aggressive breast cancer cell line MDA-MB-231 [86]. Different techniques were used to isolate the compounds: silica gel column chromatography, Sephadex LH 20, HPLC, NMR, infrared spectroscopy (IR), and tandem mass spectrometry (MS/MS).
Kim and colleagues [87] reported eight diterpenoids from the whole plant of Vitex rotundifolia L. f. (Verbenaceae). It is an important medicinal plant distributed in Korea, Japan, India, and China. The traditional uses of the plant comprise gastrointestinal diseases, chronic bronchitis, and migraine. The antiplasmodial activity was tested against the Dd2 strain, with an IC50 of 1.2 μM for the compound named abieta-11(12)-ene-9β,13β-endoperoxide, which was obtained from the n-hexane fraction (5.1 g), yielding a total of 15.4 mg. The β-peroxy bridge in this compound shows its importance for the antiplasmodial activity (Dd2), increasing it by up to 54 folds. Another promising isolated compound was Vitetrifolin E, with an IC50 1.3 μM, which was isolated with a yield of 55.7 mg from 21 g of the methanolic fraction.
Two compounds isolated from the stem barks of Trichilia monadelpha (Thonn.) (J. J. De Wilde) (Meliaceae) were very active against the P. falciparum 3D7 strain: grandifotane A showed an IC50 of 1.37 µM and khayanolide D an IC50 of 1.68 µM, both against the 3D7 strain [88]. Both isolated compounds were obtained in the n-hexane/ethyl acetate fraction (150 mg for grandifotane A and 15 mg, both from initial 150 g of extract). The genus Trichilia comprises about 70 species distributed in tropical regions, mainly Africa and America, and the species T. monadelpha is used in traditional medicine for treating a variety of diseases, including depression, epilepsy, syphilis, and rheumatism [88].
Another genus of plants, Croton, is used in Tanzanian traditional medicine for the treatment of infections, malaria, tuberculosis, and cancer. In a recent study [89], stem bark and leaf extracts of Croton kilwae Radcl.-Sm. (Euphorbiaceae) endemic species (Tanzania and Mozambique) yielded the isolation of three new crotofolane diterpenoids with antiplasmodial activity at 50 μM concentration, and which were named as crotokilwaepoxides. Artesunate at 5 μM concentration was utilized as positive control and drug vehicle DMSO as negative control in parasitaemia assays. The structure of the antiplasmodial compounds was determined via NMR spectroscopic data and mass spectrometry, and one compound was identified as a new crotofolane 9-methoxycrotokilwaepoxide A.
Still, in the terpenoids class, dimeric sesquiterpenoids isolated and characterized from Sarcandra glabra subsp. Brachystachys (Blume) (Verdc.), a plant species of tropical biome in south Asia and a long-known folk medicine in China, may also look promising in terms of antimalarial properties. Zhou and collaborators have isolated and characterized many compounds of this chemical class from plants of Chinese folk medicine [90], and, recently, it was found that one of the isolated dimeric sesquiterpenoids showed an EC50 value in the picomolar range (4.3 pM) [91], testing in vitro against P. falciparum Dd2 with DMSO as negative control. The in vitro compound has about 1000-fold potency compared to artemisinin. Therefore, this chemical class may be of particular interest in malaria drug discovery.

8. Polyphenols: Potential Antimalarial Activity

The plant Sphaerocoryne gracilis (Oliv. ex Engl. and Diels) (Verdc.) (Annonaceae) found in Tanzania was screened for antiplasmodial activity against chloroquine-sensitive (3D7) and chloroquine-resistant (Dd2) strains of P. falciparum. Compounds showing promising antiplasmodial activities, namely, (Z)-Sphaerodiol, (Z)-acetylmelodorinol, 7-hydroxy-6-hydromelodienone (13), and dichamanetin, inhibited the proliferation of P. falciparum (3D7 and Dd2), with IC50 values of 1.4–10.5 μM [92]. Silica gel column chromatography, Sephadex LH 20, HPLC, NMR (one-dimensional and two-dimensional) were employed for the isolation of compound (13). The genus Sphaerocoryne consists of only three species, namely, S. fruticosum, S. gracilis (ssp. Engleriana and gracilis), and S. punctulatum, which are native to some countries of the African continent, e.g., Angola, Zambia, Mozambique, and Tanzania [92].
Du and collaborators [93] performed a bioassay-guided isolation of the methanol extract of Galtonia regalis (Hilliard) (B.L. Burtt) (Asparagaceae), and the extract exhibited antiplasmodial activity with IC50 value less than 1.25 μg/mL. Two known cholestane glycosides and five new cholestane glycoside galtonosides were isolated after bioassay-guided fractionation. Galtonoside B (15) showed the most potent antiplasmodial activity, with an IC50 value of 214 nM against the multidrug-resistant Dd2 strain of P. falciparum. Silica gel column chromatography, Sephadex LH-20, HPLC, NMR (one-dimensional and two-dimensional), and MS/MS were used in combination for the isolation of the compound.
Phytochemical investigation revealed that 5,3′-dihydroxy-7,4′-dimethoxyflavone (1), 5,7-dihydroxy-4′-methoxyflavone (2), and 5-hydroxy-7,3′,4′-trimethoxyflavone (3) had strong antiplasmodium activity against the 3D7 strain of P. falciparum, with IC50 values of 2 nM, 5 nM, and 1 nM, respectively. The compounds were isolated from the stem bark of Drymis beccariana Gibbs (Winteraceae) and indicate a potential source of antimalarial agents [94]. Silica gel chromatography, HPLC, ultraviolet-visible spectroscopy (UV), IR, NMR, and MS/MS were used for compound isolation.
A biodereplication workflow was applied to an ethanolic extract of the medicinal plant Piper coruscans Kunth (Piperaceae) against the Plasmodium falciparum strain 3D7. This resulted in the isolation of two antimalarial chalcones, aurentiacin (7) and cardamonin (8), with IC50 values of 2.25 and 5.5 µM, respectively [95]. The combination of NMR, MS/MS and LC/MS allowed isolation and characterization of the compounds.
Dawurung and collaborators [96] identified a lead compound with a pterocarpene structure in the plant Neorautanenia mitis (A. Rich) Verdc. (Fabacae), which was developed into a drug against P. falciparum, Rautandiol B (9), with IC50 values of 400 nM (TM4/8.2) and 740 nM (K1CB1). As with the above-mentioned studies, spectroscopy techniques were utilized: UV, IR, and chromatographic techniques HPLC, Sephadex LH 20, silica gel column, and MS/MS.
In another study, the plant Aloe marlothii A.Berger (Xanthorrhoeaceae), found in southern African countries, and traditionally used in folk medicine to treat different diseases, was tested against asexual and sexual forms of P. falciparum parasites. Mianda and collaborators [97] isolated six different compounds of this plant, and the most promising compound identified was aloesaponarin I, with an IC50 of 1.54 μg/mL (5 μM) against NF54 strain and IC50 of 1.58 μg/mL (5 μM) against K1 strain.
In a recent study [98], a plant found in Asia, named Putranjiva roxburghii Wall. (Putranjivaceae), was found to contain the polyphenol quebrachitol, which had an IC50 of 0.87 μg/mL and at a dosage of 30 mg/kg indicated a chemosuppression of 73.26% in a murine model (P. berghei K173). High selectivity index was obtained for this compound (SI = 158).
The species Paeonia officinalis L. (Paeoniaceae) belongs to a genus that comprises about 33 species. They are distributed in Europe and Asia, and 11 species are found in China. The roots of P. officinalis are used in traditional Chinese medicine due to its anti-inflammatory, analgesic, and sedative properties, as well as its antihypertensive and anti-ulcer properties. Through the combination of chromatographic techniques, including Sephadex LH-20, the compounds identified as methyl gallate and galloyl paeoniflorin were isolated and tested in vitro against two different strains of P. falciparum (D6 and W2) [48], which provided better IC50 values against the W2 strain—0.61 and 2.91 µg/mL, respectively.
Antimalarial compounds from Mammea siamensis (Miq.) (T. Anders.) (Calophyllaceae) flowers, which were isolated via a bioassay-guided workflow, exhibited antimalarial activity. The result suggested that 1-hydroxy-5,6,7-trimethoxyxanthone (14) is a possible lead structure and a potent inhibitor of the PfLDH enzyme. Negative cytotoxicity in Vero and HepG2 cells was observed [70]. Techniques, such as silica column gel, HPLC, and NMR (one-dimensional and two-dimensional) were employed to isolate the antimalarial structure.

9. Conclusions and Future Perspectives

The search for antimalarial compounds isolated from plant extracts remains challenging. The benefits of searching such compounds in flora are evident: the plethora of chemical structures potentially isolated from natural sources is still an underexplored space, and the diversity in the traditional use of endemic plants also reinforces the need for isolation of possible antimalarial compounds. However, different aspects in plant extracts and their isolated compounds are crucial to be understood: the possible synergisms that exist when combining plant extracts through different techniques, the presence of factors that interfere in the composition of the extracts such as seasonality that may interfere in the levels of plants secondary metabolites, and the empirical evidence originated from the traditional use.
This mini review has provided an overview of the last five years of the literature regarding plant extracts and their isolated natural compounds with potential antimalarial effects. Twenty chemical compounds isolated from plants with IC50 < 10 μM against at least one P. falciparum strain were selected and discussed. Several rich antiplasmodial fractions derived from plant extracts have been reported in the literature. However, the link between traditional knowledge and further research in discovering potential natural products with pharmacological properties must be reinforced, and hyphenated techniques are crucial for better characterizing the chemical compounds structures and their targets.
In particular, a closer connection with the local communities and better documentation of the local scientific information gathered is needed to further aid natural products research. Some families, such as Annonaceae, Meliaceae, Asteraceae, and Euphorbiaceae, are rich sources of bioactive compounds capable of inhibiting Plasmodium in vitro. More specifically, the families Annonaceae and Asteraceae provide genera, such as Sphaerocoryn, Annickia, Petasites, and Sonchus, which could be further investigated for their antimalarial effects, based on the in vitro and in vivo experimental results, and the generation of potential lead compounds for drug development. Whether they have an appropriate cytotoxicity profile and whether they will have low IC50 values in vivo should be investigated with the use of hyphenated techniques combined with omics-based approaches.

Author Contributions

G.d.J.G.R. and S.L.R.Y. performed the literature search, analyzed data, and wrote the manuscript, G.P. wrote and revised the manuscript, C.W. revised the manuscript and supervised the work. All authors have read and agreed to the published version of the manuscript.

Funding

We are grateful for the financial support provided by the São Paulo Research Foundation (FAPESP; grant numbers 2018/18257-1, 2018/15549-1, and 2020/04923-0 [GP], 2017/26358-0 [SLRY], and 2015/26722-8 [CW]), and the doctoral fellowship by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (GJRG).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rovira-Vallbona, E.; Van Hong, N.; Kattenberg, J.H.; Huan, R.M.; Hien, N.T.T.; Ngoc, N.T.H.; Guetens, P.; Hieu, N.L.; Mai, T.T.; Duong, N.T.T.; et al. Efficacy of Dihydroartemisinin/Piperaquine and Artesunate Monotherapy for the Treatment of Uncomplicated Plasmodium falciparum Malaria in Central Vietnam. J. Antimicrob. Chemother. 2020, 75, 2272–2281. [Google Scholar] [CrossRef] [PubMed]
  2. Iwanaga, S.; Kubota, R.; Nishi, T.; Kamchonwongpaisan, S.; Srichairatanakool, S.; Shinzawa, N.; Syafruddin, D.; Yuda, M.; Uthaipibull, C. Genome-Wide Functional Screening of Drug-Resistance Genes in Plasmodium falciparum. Nat. Commun. 2022, 13, 6163. [Google Scholar] [CrossRef] [PubMed]
  3. World Health Organization. World Malaria Report 2022; World Health Organization: Geneva, Switzerland, 2022.
  4. Datoo, M.S.; Natama, H.M.; Somé, A.; Bellamy, D.; Traoré, O.; Rouamba, T.; Tahita, M.C.; Ido, N.F.A.; Yameogo, P.; Valia, D.; et al. Efficacy and Immunogenicity of R21/Matrix-M Vaccine against Clinical Malaria after 2 Years’ Follow-up in Children in Burkina Faso: A Phase 1/2b Randomised Controlled Trial. Lancet Infect. Dis. 2022, 22, 1728–1736. [Google Scholar] [CrossRef] [PubMed]
  5. Kassegne, K.; Komi Koukoura, K.; Shen, H.M.; Chen, S.B.; Fu, H.T.; Chen, Y.Q.; Zhou, X.N.; Chen, J.H.; Cheng, Y. Genome-Wide Analysis of the Malaria Parasite Plasmodium falciparum Isolates From Togo Reveals Selective Signals in Immune Selection-Related Antigen Genes. Front. Immunol. 2020, 11, 552698. [Google Scholar] [CrossRef] [PubMed]
  6. Mulenga, M.C.; Sitali, L.; Ciubotariu, I.I.; Hawela, M.B.; Hamainza, B.; Chipeta, J.; Mharakurwa, S. Decreased Prevalence of the Plasmodium falciparum Pfcrt K76T and Pfmdr1 and N86Y Mutations Post-Chloroquine Treatment Withdrawal in Katete District, Eastern Zambia. Malar. J. 2021, 20, 329. [Google Scholar] [CrossRef] [PubMed]
  7. Russo, G.; L’Episcopia, M.; Menegon, M.; Souza, S.S.; Dongho, B.G.D.; Vullo, V.; Lucchi, N.W.; Severini, C. Dihydroartemisinin–Piperaquine Treatment Failure in Uncomplicated Plasmodium falciparum Malaria Case Imported from Ethiopia. Infection 2018, 46, 867–870. [Google Scholar] [CrossRef]
  8. Hassen, J.; Alemayehu, G.S.; Dinka, H.; Golassa, L. High Prevalence of Pfcrt 76T and Pfmdr1 N86 Genotypes in Malaria Infected Patients Attending Health Facilities in East Shewa Zone, Oromia Regional State, Ethiopia. Malar. J. 2022, 21, 286. [Google Scholar] [CrossRef]
  9. Froeschl, G.; Saathoff, E.; Kroidl, I.; Berens-Riha, N.; Clowes, P.; Maboko, L.; Assisya, W.; Mwalongo, W.; Gerhardt, M.; Ntinginya, E.N.; et al. Reduction of Malaria Prevalence after Introduction of Artemisinin-Combination-Therapy in Mbeya Region, Tanzania: Results from a Cohort Study with 6773 Participants. Malar. J. 2018, 17, 245. [Google Scholar] [CrossRef]
  10. Villena, F.E.; Sanchez, J.F.; Nolasco, O.; Braga, G.; Ricopa, L.; Barazorda, K.; Salas, C.J.; Lucas, C.; Lizewski, S.E.; Joya, C.A.; et al. Drug Resistance and Population Structure of Plasmodium falciparum and Plasmodium vivax in the Peruvian Amazon. Sci. Rep. 2022, 12, 16474. [Google Scholar] [CrossRef]
  11. Su, X.-Z.; Miller, L.H. The Discovery of Artemisinin and the Nobel Prize in Physiology or Medicine. Sci. China Life Sci. 2015, 58, 1175–1179. [Google Scholar] [CrossRef]
  12. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed]
  13. Thanh, N.V.; Thuy-Nhien, N.; Tuyen, N.T.K.; Tong, N.T.; Nha-Ca, N.T.; Dong, L.T.; Quang, H.H.; Farrar, J.; Thwaites, G.; White, N.J.; et al. Rapid Decline in the Susceptibility of Plasmodium falciparum to Dihydroartemisinin–Piperaquine in the South of Vietnam. Malar. J. 2017, 16, 27. [Google Scholar] [CrossRef] [PubMed]
  14. Pernicová, E.; Krsek, M. Current Trends in the Epidemiology of Malaria. Epidemiol. Mikrobiol. Imunol. 2022, 71, 175–178. [Google Scholar] [PubMed]
  15. Moxon, C.A.; Gibbins, M.P.; Mcguinness, D.; Milner, D.A.; Marti, M. New Insights into Malaria Pathogenesis. Annu. Rev. Pathol. 2020, 15, 315–343. [Google Scholar] [CrossRef]
  16. Ministério da Saúde. Boletim Epidemiológico Vol.53 No 17. Available online: https://www.gov.br/saude/pt-br/centrais-de-conteudo/publicacoes/boletins/epidemiologicos/edicoes/2022/boletim-epidemiologico-vol-53-no17.pdf/view (accessed on 6 March 2023).
  17. Lindblade, K.A.; Li Xiao, H.; Tiffany, A.; Galappaththy, G.; Alonso, P.; Abeyasinghe, R.; Akpaka, K.; Aragon-Lopez, M.A.; Baba, E.S.; Bahena, A.; et al. Supporting Countries to Achieve Their Malaria Elimination Goals: The WHO E-2020 Initiative. Malar. J. 2021, 20, 481. [Google Scholar] [CrossRef]
  18. Pan, W.-H.; Xu, X.-Y.; Shi, N.; Tsang, S.; Zhang, H.-J. Antimalarial Activity of Plant Metabolites. Int. J. Mol. Sci. 2018, 19, 1382. [Google Scholar] [CrossRef]
  19. Kaur, H.; Mukhtar, H.M.; Singh, A.; Mahajan, A. Antiplasmodial Medicinal Plants: A Literature Review on Efficacy, Selectivity and Phytochemistry of Crude Plant Extracts. J. Biol. Act. Prod. Nat. 2018, 8, 272–294. [Google Scholar] [CrossRef]
  20. Cock, I.E.; Selesho, M.I.; van Vuuren, S.F. A Review of the Traditional Use of Southern African Medicinal Plants for the Treatment of Malaria. J. Ethnopharmacol. 2019, 245, 112176. [Google Scholar] [CrossRef]
  21. Noronha, M.; Pawar, V.; Prajapati, A.; Subramanian, R.B. A Literature Review on Traditional Herbal Medicines for Malaria. S. Afr. J. Bot. 2020, 128, 292–303. [Google Scholar] [CrossRef]
  22. Bekono, B.D.; Ntie-Kang, F.; Onguéné, P.A.; Lifongo, L.L.; Sippl, W.; Fester, K.; Owono, L.C.O. The Potential of Anti-Malarial Compounds Derived from African Medicinal Plants: A Review of Pharmacological Evaluations from 2013 to 2019. Malar. J. 2020, 19, 183. [Google Scholar] [CrossRef]
  23. Nigussie, G.; Wale, M. Medicinal Plants Used in Traditional Treatment of Malaria in Ethiopia: A Review of Ethnomedicine, Anti-Malarial and Toxicity Studies. Malar. J. 2022, 21, 262. [Google Scholar] [CrossRef] [PubMed]
  24. Moyo, P.; Mugumbate, G.; Eloff, J.N.; Louw, A.I.; Maharaj, V.J.; Birkholtz, L.M. Natural Products: A Potential Source of Malaria Transmission Blocking Drugs? Pharmaceuticals 2020, 13, 251. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, X.; Wang, Y.; Ma, N.; Tian, J.; Shao, Y.; Zhu, B.; Wong, Y.K.; Liang, Z.; Zou, C.; Wang, J. Target Identification of Natural Medicine with Chemical Proteomics Approach: Probe Synthesis, Target Fishing and Protein Identification. Signal Transduct. Target. Ther. 2020, 5, 72. [Google Scholar] [CrossRef] [PubMed]
  26. Kam, C.M.; Abuelyaman, A.S.; Li, Z.; Hudig, D.; Powers, J.C. Biotinylated Isocoumarins, New Inhibitors and Reagents for Detection, Localization, and Isolation of Serine Proteases. Bioconjug. Chem. 1993, 4, 560–567. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, Y.; Patricelli, M.P.; Cravatt, B.F. Activity-Based Protein Profiling: The Serine Hydrolases. Proc. Natl. Acad. Sci. USA 1999, 96, 14694–14699. [Google Scholar] [CrossRef] [PubMed]
  28. Murale, D.P.; Hong, S.C.; Haque, M.M.; Lee, J.-S. Photo-Affinity Labeling (PAL) in Chemical Proteomics: A Handy Tool to Investigate Protein-Protein Interactions (PPIs). Proteome Sci. 2016, 15, 14. [Google Scholar] [CrossRef] [PubMed]
  29. Molina, D.M.; Jafari, R.; Ignatushchenko, M.; Seki, T.; Larsson, E.A.; Dan, C.; Sreekumar, L.; Cao, Y.; Nordlund, P. Monitoring Drug Target Engagement in Cells and Tissues Using the Cellular Thermal Shift Assay. Science 2013, 341, 84–87. [Google Scholar] [CrossRef]
  30. Gaetani, M.; Sabatier, P.; Saei, A.A.; Beusch, C.M.; Yang, Z.; Lundström, S.L.; Zubarev, R.A. Proteome Integral Solubility Alteration: A High-Throughput Proteomics Assay for Target Deconvolution. J. Proteome Res. 2019, 18, 4027–4037. [Google Scholar] [CrossRef]
  31. Dziekan, J.M.; Yu, H.; Chen, D.; Dai, L.; Wirjanata, G.; Larsson, A.; Prabhu, N.; Sobota, R.M.; Bozdech, Z.; Nordlund, P. Identifying Purine Nucleoside Phosphorylase as the Target of Quinine Using Cellular Thermal Shift Assay. Sci. Transl. Med. 2019, 11, eaau3174. [Google Scholar] [CrossRef]
  32. Lomenick, B.; Hao, R.; Jonai, N.; Chin, R.M.; Aghajan, M.; Warburton, S.; Wang, J.; Wu, R.P.; Gomez, F.; Loo, J.A.; et al. Target Identification Using Drug Affinity Responsive Target Stability (DARTS). Proc. Natl. Acad. Sci. USA 2009, 106, 21984–21989. [Google Scholar] [CrossRef]
  33. Piazza, I.; Kochanowski, K.; Cappelletti, V.; Fuhrer, T.; Noor, E.; Sauer, U.; Picotti, P. A Map of Protein-Metabolite Interactions Reveals Principles of Chemical Communication. Cell 2018, 172, 358–372.e23. [Google Scholar] [CrossRef] [PubMed]
  34. West, G.M.; Tang, L.; Fitzgerald, M.C. Thermodynamic Analysis of Protein Stability and Ligand Binding Using a Chemical Modification- and Mass Spectrometry-Based Strategy. Anal. Chem. 2008, 80, 4175–4185. [Google Scholar] [CrossRef] [PubMed]
  35. Van Vranken, J.G.; Li, J.; Mitchell, D.C.; Navarrete-Perea, J.; Gygi, S.P. Assessing Target Engagement Using Proteome-Wide Solvent Shift Assays. eLife 2021, 10, e70784. [Google Scholar] [CrossRef] [PubMed]
  36. Yen, K.; Travins, J.; Wang, F.; David, M.D.; Artin, E.; Straley, K.; Padyana, A.; Gross, S.; DeLaBarre, B.; Tobin, E.; et al. AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations. Cancer Discov. 2017, 7, 478–493. [Google Scholar] [CrossRef] [PubMed]
  37. Popovici-Muller, J.; Lemieux, R.M.; Artin, E.; Saunders, J.O.; Salituro, F.G.; Travins, J.; Cianchetta, G.; Cai, Z.; Zhou, D.; Cui, D.; et al. Discovery of AG-120 (Ivosidenib): A First-in-Class Mutant IDH1 Inhibitor for the Treatment of IDH1 Mutant Cancers. ACS Med. Chem. Lett. 2018, 9, 300–305. [Google Scholar] [CrossRef] [PubMed]
  38. Ward, P.S.; Patel, J.; Wise, D.R.; Abdel-Wahab, O.; Bennett, B.D.; Coller, H.A.; Cross, J.R.; Fantin, V.R.; Hedvat, C.V.; Perl, A.E.; et al. The Common Feature of Leukemia-Associated IDH1 and IDH2 Mutations Is a Neomorphic Enzyme Activity Converting α-Ketoglutarate to 2-Hydroxyglutarate. Cancer Cell 2010, 17, 225–234. [Google Scholar] [CrossRef]
  39. Dang, L.; White, D.W.; Gross, S.; Bennett, B.D.; Bittinger, M.A.; Driggers, E.M.; Fantin, V.R.; Jang, H.G.; Jin, S.; Keenan, M.C.; et al. Cancer-Associated IDH1 Mutations Produce 2-Hydroxyglutarate. Nature 2009, 462, 739–744. [Google Scholar] [CrossRef]
  40. Freire, R.T.; Bero, J.; Beaufay, C.; Selegato, D.M.; Coqueiro, A.; Choi, Y.H.; Quetin-Leclercq, J. Identification of Antiplasmodial Triterpenes from Keetia Species Using NMR-Based Metabolic Profiling. Metabolomics 2019, 15, 27. [Google Scholar] [CrossRef]
  41. Graziani, V.; Scognamiglio, M.; Belli, V.; Esposito, A.; D’Abrosca, B.; Chambery, A.; Russo, R.; Panella, M.; Russo, A.; Ciardiello, F.; et al. Metabolomic Approach for a Rapid Identification of Natural Products with Cytotoxic Activity against Human Colorectal Cancer Cells. Sci. Rep. 2018, 8, 5309. [Google Scholar] [CrossRef]
  42. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; International Natural Product Sciences Taskforce; Supuran, C.T. Natural Products in Drug Discovery: Advances and Opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  43. Hubert, J.; Nuzillard, J.-M.; Renault, J.-H. Dereplication Strategies in Natural Product Research: How Many Tools and Methodologies behind the Same Concept? Phytochem. Rev. 2017, 16, 55–95. [Google Scholar] [CrossRef]
  44. Drennan, B.W.; Wicker, A.P.; Berger, B.K.; Schug, K.A. Measurements of Drugs and Metabolites in Biological Matrices Using SFC and SFE-SFC-MS. In Practical Application of Supercritical Fluid Chromatography for Pharmaceutical Research and Development; Elsevier: Amsterdam, The Netherlands, 2022; pp. 73–99. [Google Scholar]
  45. Ketteler, R.; Kriston-Vizi, J. High-Content Screening in Cell Biology. In Encyclopedia of Cell Biology; Elsevier: Amsterdam, The Netherlands, 2016; pp. 234–244. [Google Scholar]
  46. Li, Z.; Cvijic, M.E.; Zhang, L. Cellular Imaging in Drug Discovery: Imaging and Informatics for Complex Cell Biology. In Comprehensive Medicinal Chemistry III; Elsevier: Amsterdam, The Netherlands, 2017; pp. 362–387. [Google Scholar]
  47. Wolfender, J.-L.; Litaudon, M.; Touboul, D.; Queiroz, E.F. Innovative Omics-Based Approaches for Prioritisation and Targeted Isolation of Natural Products—New Strategies for Drug Discovery. Nat. Prod. Rep. 2019, 36, 855–868. [Google Scholar] [CrossRef] [PubMed]
  48. Kurita, K.L.; Linington, R.G. Connecting Phenotype and Chemotype: High-Content Discovery Strategies for Natural Products Research. J. Nat. Prod. 2015, 78, 587–596. [Google Scholar] [CrossRef] [PubMed]
  49. Mbouna, C.D.J.; Kouipou, R.M.T.; Keumoe, R.; Tchokouaha, L.R.Y.; Fokou, P.V.T.; Tali, B.M.T.; Sahal, D.; Boyom, F.F. Potent Antiplasmodial Extracts and Fractions from Terminalia mantaly and Terminalia superba. Malar. J. 2018, 17, 142. [Google Scholar] [CrossRef]
  50. Chibli, L.A.; Rosa, A.L.; Nonato, M.C.; Da Costa, F.B. Untargeted LC–MS Metabolomic Studies of Asteraceae Species to Discover Inhibitors of Leishmania Major Dihydroorotate Dehydrogenase. Metabolomics 2019, 15, 59. [Google Scholar] [CrossRef]
  51. Navarro-Muñoz, J.C.; Selem-Mojica, N.; Mullowney, M.W.; Kautsar, S.A.; Tryon, J.H.; Parkinson, E.I.; De Los Santos, E.L.C.; Yeong, M.; Cruz-Morales, P.; Abubucker, S.; et al. A Computational Framework to Explore Large-Scale Biosynthetic Diversity. Nat. Chem. Biol. 2020, 16, 60–68. [Google Scholar] [CrossRef]
  52. Wang, M.; Carver, J.J.; Phelan, V.V.; Sanchez, L.M.; Garg, N.; Peng, Y.; Nguyen, D.D.; Watrous, J.; Kapono, C.A.; Luzzatto-Knaan, T.; et al. Sharing and Community Curation of Mass Spectrometry Data with Global Natural Products Social Molecular Networking. Nat. Biotechnol. 2016, 34, 828–837. [Google Scholar] [CrossRef]
  53. Xu, J.; Wei, K.; Zhang, G.; Lei, L.; Yang, D.; Wang, W.; Han, Q.; Xia, Y.; Bi, Y.; Yang, M.; et al. Ethnopharmacology, Phytochemistry, and Pharmacology of Chinese Salvia Species: A Review. J. Ethnopharmacol. 2018, 225, 18–30. [Google Scholar] [CrossRef]
  54. Krettli, A.U.; Adebayo, J.O.; Krettli, L.G. Testing of Natural Products and Synthetic Molecules Aiming at New Antimalarials. Curr. Drug Targets 2009, 10, 261–270. [Google Scholar] [CrossRef]
  55. Wein, S.; Maynadier, M.; Van Ba, C.T.; Cerdan, R.; Peyrottes, S.; Fraisse, L.; Vial, H. Reliability of Antimalarial Sensitivity Tests Depends on Drug Mechanisms of Action. J. Clin. Microbiol. 2010, 48, 1651–1660. [Google Scholar] [CrossRef]
  56. Mokgethi-Morule, T.; N’Da, D.D. Cell Based Assays for Anti-Plasmodium Activity Evaluation. Eur. J. Pharm. Sci. 2016, 84, 26–36. [Google Scholar] [CrossRef] [PubMed]
  57. Abdulrazak, A. In Vitro Antimalarial Activity of Extracts of Some Indigenous Plant Species in Kebbi State. UMYU J. Microbiol. Res. 2020, 5, 1–10. [Google Scholar] [CrossRef]
  58. Boeno, S.I.; Vieira, I.J.C.; Braz-Filho, R.; de Souza Passos, M.; Curcino Vieira, M.G.; do Nascimento, M.F.A.; Gontijo, D.C.; de Oliveira, A.B. Antiplasmodial and Cytotoxic Effects of the Methanol Extract, Canthinone Alkaloids, Squalene- and Protolimonoid-Type Triterpenes from Homalolepis suffruticosa Roots. J. Ethnopharmacol. 2022, 285, 114890. [Google Scholar] [CrossRef]
  59. Kaharudin, F.A.; Zohdi, R.M.; Mukhtar, S.M.; Sidek, H.M.; Bihud, N.V.; Rasol, N.E.; Ahmad, F.B.; Ismail, N.H. In Vitro Antiplasmodial and Cytotoxicity Activities of Crude Extracts and Major Compounds from Goniothalamus lanceolatus. J. Ethnopharmacol. 2020, 254, 112657. [Google Scholar] [CrossRef]
  60. Dwivedi, M.K.; Shukla, R.; Sharma, N.K.; Manhas, A.; Srivastava, K.; Kumar, N.; Singh, P.K. Evaluation of Ethnopharmacologically Selected Vitex negundo L. for In Vitro Antimalarial Activity and Secondary Metabolite Profiling. J. Ethnopharmacol. 2021, 275, 114076. [Google Scholar] [CrossRef]
  61. Yun, H.S.; Dinzouna-Boutamba, S.D.; Lee, S.; Moon, Z.; Kwak, D.; Chung, D.I.; Hong, Y.; Rhee, M.H.; Goo, Y.K. Petasites Japonicus Extract Exerts Anti-Malarial Effects by Inhibiting Platelet Activation. Phytomedicine 2022, 102, 154167. [Google Scholar] [CrossRef] [PubMed]
  62. Pratama, A.N.W.; Pratoko, D.K.; Triatmoko, B.; Winarto, N.B.; Laksono, T.A.; Nugraha, A.S. Senna occidentalis and Cyanthillium patulum: Indonesian Herbs as Source for Antimalarial Agents. Proc. Int. Conf. Green Technol. 2019, 10, 9. [Google Scholar] [CrossRef]
  63. Budiarti, M.; Maruzy, A.; RK, N.R.; Brotojoyo, E. Aktivitas Antimalaria Daun Gempol (Nauclea orientalis (L.) L) Terhadap Plasmodium falciparum. Media Penelit. Pengemb. Kesehat. 2020, 30, 135–146. [Google Scholar] [CrossRef]
  64. Cudjoe, E.; Cudjoe, E.; Donu, D.; Donu, D.; Okonu, R.E.; Okonu, R.E.; Amponsah, J.A.; Amoah, L.E.; Amoah, L.E. The in Vitro Antiplasmodial Activities of Aqueous Extracts of Selected Ghanaian Herbal Plants. J. Parasitol. Res. 2020, 2020, 5041919. [Google Scholar] [CrossRef]
  65. Ekasari, W.; Widya Pratiwi, D.; Amanda, Z.; Suciati; Widyawaruyanti, A.; Arwati, H.; Uzor, P.F. Various Parts of Helianthus annuus Plants as New Sources of Antimalarial Drugs. Evid.-Based Complement. Altern. Med. 2019, 2019, 7390385. [Google Scholar] [CrossRef]
  66. Koffi, J.A.; Silué, K.D.; Tano, D.K.; Dable, T.M.; Yavo, W. Evaluation of Antiplasmodial Activity of Extracts from Endemic Medicinal Plants Used to Treat Malaria in Côte d’Ivoire. BioImpacts 2020, 10, 151–157. [Google Scholar] [CrossRef] [PubMed]
  67. Sevik Kilicaslan, O.; Cretton, S.; Quirós-Guerrero, L.; Bella, M.A.; Kaiser, M.; Mäser, P.; Ndongo, J.T.; Cuendet, M. Isolation and Structural Elucidation of Compounds from Pleiocarpa bicarpellata and Their In Vitro Antiprotozoal Activity. Molecules 2022, 27, 2200. [Google Scholar] [CrossRef] [PubMed]
  68. Gontijo, D.C.; do Nascimento, M.F.A.; Rody, H.V.S.; Magalhães, R.A.; Margalho, L.F.; Brandão, G.C.; de Oliveira, A.B. In Vitro Antiplasmodial Activity, Targeted LC–MS Metabolite Profiling, and Identification of Major Natural Products in the Bioactive Extracts of Palicourea and Psychotria Species from the Amazonia and Atlantic Forest Biomes, Brazil. Metabolomics 2021, 17, 81. [Google Scholar] [CrossRef]
  69. Dwivedi, M.K.; Mishra, S.; Sonter, S.; Singh, P.K. Diterpenoids as Potential Anti-Malarial Compounds from Andrographis paniculata. Beni-Suef Univ. J. Basic Appl. Sci. 2021, 10, 7. [Google Scholar] [CrossRef]
  70. Chaniad, P.; Techarang, T.; Phuwajaroanpong, A.; Horata, N.; Septama, A.W.; Punsawad, C. Exploring Potential Antimalarial Candidate from Medicinal Plants of Kheaw Hom Remedy. Trop. Med. Infect. Dis. 2022, 7, 368. [Google Scholar] [CrossRef] [PubMed]
  71. Wahyuni, D.K.; Wacharasindhu, S.; Bankeeree, W.; Wahyuningsih, S.P.A.; Ekasari, W.; Purnobasuki, H.; Punnapayak, H.; Prasongsuk, S. In Vitro and in Vivo Antiplasmodial Activities of Leaf Extracts from Sonchus arvensis L. BMC Complement. Med. Ther. 2023, 23, 47. [Google Scholar] [CrossRef] [PubMed]
  72. Chaniad, P.; Techarang, T.; Phuwajaroanpong, A.; Plirat, W.; Viriyavejakul, P.; Septama, A.W.; Punsawad, C. Antimalarial Efficacy and Toxicological Assessment of Medicinal Plant Ingredients of Prabchompoothaweep Remedy as a Candidate for Antimalarial Drug Development. BMC Complement. Med. Ther. 2023, 23, 12. [Google Scholar] [CrossRef]
  73. Rummun, N.; Payne, B.; Blom van Staden, A.; Twilley, D.; Houghton, B.; Horrocks, P.; Li, W.-W.; Lall, N.; Bahorun, T.; Neergheen, V.S. Pluripharmacological Potential of Mascarene Endemic Plant Leaf Extracts. Biocatal. Agric. Biotechnol. 2023, 47, 102572. [Google Scholar] [CrossRef]
  74. Djouwoug, C.N.; Gounoue, R.K.; Ngueguim, F.T.; NankapTsakem, J.M.; Gouni, C.D.; Kandeda, A.C.; Ngouela, S.; Lenta, B.N.; Sewald, N.; Fekam, F.B.; et al. In Vitro and in Vivo Antiplasmodial Activity of Hydroethanolic Bark Extract of Bridelia atroviridis Müll. Arg. (Euphorbiaceae) and Lc-Ms-Based Phytochemical Analysis. J. Ethnopharmacol. 2021, 266, 113424. [Google Scholar] [CrossRef]
  75. Erhunse, N.; Omoregie, E.S.; Sahal, D. Antiplasmodial and Antimalarial Evaluation of a Nigerian Hepta-Herbal Agbo-Iba Decoction: Identification of Magic Bullets and Possible Facilitators of Drug Action. J. Ethnopharmacol. 2023, 301, 115807. [Google Scholar] [CrossRef]
  76. Guo, X.; Tang, C.C.; Thomas, D.C.; Couvreur, T.L.P.; Saunders, R.M.K. A Mega-Phylogeny of the Annonaceae: Taxonomic Placement of Five Enigmatic Genera and Support for a New Tribe, Phoenicantheae. Sci. Rep. 2017, 7, 7323. [Google Scholar] [CrossRef] [PubMed]
  77. Panda, S.K.; Luyten, W. Antiparasitic Activity in Asteraceae with Special Attention to Ethnobotanical Use by the Tribes of Odisha, India. Parasite 2018, 25, 10. [Google Scholar] [CrossRef] [PubMed]
  78. Mogaka, S.; Molu, H.; Kagasi, E.; Ogila, K.; Waihenya, R.; Onditi, F.; Ozwara, H. Senna occidentalis (L.) Link Root Extract Inhibits Plasmodium Growth in Vitro and in Mice. BMC Complement. Med. Ther. 2023, 23, 71. [Google Scholar] [CrossRef] [PubMed]
  79. Ledoux, A.; Hamann, C.; Bonnet, O.; Jullien, K.; Quetin-Leclercq, J.; Tchinda, A.; Smadja, J.; Gauvin-Bialecki, A.; Maquoi, E.; Frédérich, M. Bioactive Clerodane Diterpenoids from the Leaves of Casearia coriacea Vent. Molecules 2023, 28, 1197. [Google Scholar] [CrossRef] [PubMed]
  80. Abdel-ٍٍSattar, E.; Abdallah, H.M.; El-Mekkawy, S.; Ichino, C.; Kiyohara, H.; Yamada, H. Antimalarial Alkaloid from Hypoestes Forskaolii. Exp. Parasitol. 2020, 211, 107851. [Google Scholar] [CrossRef]
  81. Bhatt, V.; Kumari, S.; Upadhyay, P.; Agrawal, P.; Anmol; Sahal, D.; Sharma, U. Chemical Profiling and Quantification of Potential Active Constituents Responsible for the Antiplasmodial Activity of Cissampelos Pareira. J. Ethnopharmacol. 2020, 262, 113185. [Google Scholar] [CrossRef] [PubMed]
  82. Anmol; Kumari, S.; Kumar, R.; Singh, R.; Aggarwal, G.; Agrawal, P.; Sahal, D.; Sharma, U. Antiplasmodial Diterpenoid Alkaloid from Aconitum heterophyllum Wall. Ex Royle: Isolation, Characterization, and UHPLC-DAD Based Quantification. J. Ethnopharmacol. 2022, 287, 114931. [Google Scholar] [CrossRef]
  83. Keita, A.; Franetich, J.-F.; Carraz, M.; Valentin, L.; Bordessoules, M.; Baron, L.; Bigeard, P.; Dupuy, F.; Geay, V.; Tefit, M.; et al. Potent Antiplasmodial Derivatives of Dextromethorphan Reveal the Ent-Morphinan Pharmacophore of Tazopsine-Type Alkaloids. Pharmaceutics 2022, 14, 372. [Google Scholar] [CrossRef]
  84. Greve, H.L.; Kaiser, M.; Kaiser, M.; Schmidt, T.J. Investigation of Antiplasmodial Effects of Terpenoid Compounds Isolated from Myrrh. Planta Med. 2020, 86, 643–654. [Google Scholar] [CrossRef]
  85. Baldé, M.A.; Tuenter, E.; Traoré, M.S.; Peeters, L.; Matheeussen, A.; Cos, P.; Caljon, G.; Vermeyen, T.; Herrebout, W.; Balde, A.M.; et al. Antiplasmodial Oleanane Triterpenoids from Terminalia albida Root Bark. J. Nat. Prod. 2021, 84, 666–675. [Google Scholar] [CrossRef]
  86. Nyandoro, S.S.; Maeda, G.; Munissi, J.J.E.; Gruhonjic, A.; Fitzpatrick, P.A.; Lindblad, S.; Duffy, S.; Pelletier, J.; Pan, F.; Puttreddy, R.; et al. A New Benzopyranyl Cadenane Sesquiterpene and Other Antiplasmodial and Cytotoxic Metabolites from Cleistochlamys kirkii. Molecules 2019, 24, 2746. [Google Scholar] [CrossRef] [PubMed]
  87. Kim, Y.A.; Latif, A.; Kong, C.-S.; Seo, Y.; Dalal, S.R.; Cassera, M.B.; Kingston, D.G.I. Antimalarial Diterpenoids from Vitex Rotundifolia: Isolation, Structure Elucidation, and in Vitro Antiplasmodial Activity. Bioorg. Chem. 2020, 100, 103925. [Google Scholar] [CrossRef] [PubMed]
  88. Djoumessi, A.K.; Nono, R.N.; Neumann, B.; Stammler, H.-G.; Bitchagno, G.T.M.; Efange, N.M.; Nkenfou, C.N.; Ayong, L.; Lenta, B.N.; Sewald, N.; et al. Constituents of the Stem Bark of Trichilia monadelpha (Thonn.) J. J. De Wilde (Meliaceae) and Their Antibacterial and Antiplasmodial Activities. Metabolites 2023, 13, 298. [Google Scholar] [CrossRef] [PubMed]
  89. Mahambo, E.T.; Uwamariya, C.; Miah, M.; da Costa Clementino, L.; Alvarez, L.C.S.; Di Santo Meztler, G.P.; Trybala, E.; Said, J.; Wieske, L.H.E.; Ward, J.S.; et al. Crotofolane Diterpenoids and Other Constituents Isolated from Croton kilwae. J. Nat. Prod. 2023, 86, 380–389. [Google Scholar] [CrossRef] [PubMed]
  90. Zhou, B.; Wu, Y.; Dalal, S.; Merino, E.F.; Liu, Q.-F.; Xu, C.-H.; Yuan, T.; Ding, J.; Kingston, D.G.I.; Cassera, M.B.; et al. Nanomolar Antimalarial Agents against Chloroquine-Resistant Plasmodium falciparum from Medicinal Plants and Their Structure–Activity Relationships. J. Nat. Prod. 2017, 80, 96–107. [Google Scholar] [CrossRef]
  91. Zhou, B.; Zimbres, F.M.; Butler, J.H.; Xu, C.H.; Haney, R.S.; Wu, Y.; Cassera, M.B.; Yue, J.M. Picomolar Antimalarial Agent from a Chinese Medicinal Plant. Sci. China Chem. 2022, 65, 82–86. [Google Scholar] [CrossRef]
  92. Maeda, G.; Munissi, J.J.E.; Lindblad, S.; Duffy, S.; Pelletier, J.; Avery, V.M.; Nyandoro, S.S.; Erdélyi, M. A Meroisoprenoid, Heptenolides, and C-Benzylated Flavonoids from Sphaerocoryne gracilis ssp. Gracilis. J. Nat. Prod. 2020, 83, 316–322. [Google Scholar] [CrossRef]
  93. Du, Y.; Martin, B.A.; Valenciano, A.L.; Clement, J.A.; Goetz, M.; Cassera, M.B.; Kingston, D.G.I. Galtonosides A-E: Antiproliferative and Antiplasmodial Cholestane Glycosides from Galtonia regalis. J. Nat. Prod. 2020, 83, 1043–1050. [Google Scholar] [CrossRef]
  94. Herlina, T.; Rudiana, T.; Julaeha, E.; Parubak, A.S. Flavonoids from Stem Bark of Akway (Drymis beccariana Gibs) and Theirs Antimalarial Properties. In Proceedings of the Journal of Physics: Conference Series; Institute of Physics Publishing: Bristol, UK, 2019; Volume 1280. [Google Scholar]
  95. Vásquez-Ocmín, P.G.; Gallard, J.-F.; Van Baelen, A.-C.; Leblanc, K.; Cojean, S.; Mouray, E.; Grellier, P.; Guerra, C.A.A.; Beniddir, M.A.; Evanno, L.; et al. Biodereplication of Antiplasmodial Extracts: Application of the Amazonian Medicinal Plant Piper coruscans Kunth. Molecules 2022, 27, 7638. [Google Scholar] [CrossRef]
  96. Dawurung, C.J.; Nguyen, M.T.H.; Pengon, J.; Dokladda, K.; Bunyong, R.; Rattanajak, R.; Kamchonwongpaisan, S.; Nguyen, P.T.M.; Pyne, S.G. Isolation of Bioactive Compounds from Medicinal Plants Used in Traditional Medicine: Rautandiol B, a Potential Lead Compound against Plasmodium falciparum. BMC Complement. Med. Ther. 2021, 21, 231. [Google Scholar] [CrossRef]
  97. Mianda, S.M.; Invernizzi, L.; van der Watt, M.E.; Reader, J.; Moyo, P.; Birkholtz, L.-M.; Maharaj, V.J. In Vitro Dual Activity of Aloe Marlothii Roots and Its Chemical Constituents against Plasmodium falciparum Asexual and Sexual Stage Parasites. J. Ethnopharmacol. 2022, 297, 115551. [Google Scholar] [CrossRef] [PubMed]
  98. Mishra, S.; Kumar, S.; Ramdas; Khare, S.; Shukla, A.; Shanker, K.; Pal, A.; Khan, F.; Darokar, M.P. Quebrachitol from Putranjiva roxburghii Wall. (Putranjivaceae) a Potent Antimalarial: Pre-Clinical Efficacy and Its Interaction with PfLDH. Parasitol. Int. 2023, 92, 102675. [Google Scholar] [CrossRef] [PubMed]
Figure 1. General workflow for discovery and isolation of chemical compounds derived from natural products. The figure was generated with the help of BioRender.com (License #2364–1511, Toronto, ON, Canada).
Figure 1. General workflow for discovery and isolation of chemical compounds derived from natural products. The figure was generated with the help of BioRender.com (License #2364–1511, Toronto, ON, Canada).
Pharmaceutics 15 01638 g001
Figure 2. Isolated and characterized chemical compounds from different plant sources that showed low IC50 (<10 μM).
Figure 2. Isolated and characterized chemical compounds from different plant sources that showed low IC50 (<10 μM).
Pharmaceutics 15 01638 g002aPharmaceutics 15 01638 g002bPharmaceutics 15 01638 g002c
Table 1. Selection of promising enriched extracts against P. falciparum in vitro. Chloroquine-resistant strains: W2, K1, W2, Cam WT_C580Y, and IPC4912. Chloroquine-sensitive strains: 3D7 and NF54.
Table 1. Selection of promising enriched extracts against P. falciparum in vitro. Chloroquine-resistant strains: W2, K1, W2, Cam WT_C580Y, and IPC4912. Chloroquine-sensitive strains: 3D7 and NF54.
Plant SpeciesPlant PartsType of ExtractIC50 Value against P. falciparum (μg/mL)References
Azadirachta indica (A. Juss)LeavesEthanol | Aqueous7.4 (ethanol) and 8.6 (aqueous)[57]
Homalolepis suffruticosa (Engl.) (Devecchi and Pirani)RootsMethanol1.88 (W2)[58]
Goniothalamus
Lanceolatus (Miq.)
RootsMethanol2.7 (3D7) and 1.7 (K1)[59]
Vitex negundo (L.)LeavesChloroform7.21 (3D7) and 7.43 μg/mL (K1)[60]
Petasites japonicus (Siebold and Zucc.) (Maxim)LeavesEthanol (70%)8.48 (3D7) and 7.83 (Dd2)[61]
Senna occidentalis (L.) (Link)Leaves and seedsMethanol12.19 (leaves) and 6.82 (seeds)[62]
RootsMethanol1.76 (3D7)
Nauclea orientalis (L.)LeavesMethanol3.91 (3D7)[63]
Alchornea cordifolia (Schumach.) (Müll. Arg.)HerbalAqueous5.8 (NF54), 17.4 (Cam WT_C580Y), and 15.8 (IPC 4912)[64]
Helianthus annuus (L.)Roots and leavesEthanol2.3 (3D7)[65]
Harungana
madagascariensis Lam. ex Poir.
BarksAqueous6.16 (NF54)[66]
Pericopsis laxiflora (Benth. ex Baker) (Meeuwen)LeavesMethanol7.44 (K1)[66]
Pleiocarpa bicarpellate (Stapf)RootsDichloromethane/Methanol (1:1)3.5 (NF54)[67]
Psychotria apoda (Steyerm.) (Delprete and J.H.Kirkbr.)LeavesAcid-base>90% (W2)[68]
Andrographis paniculate (Burm.f.) (Wall.)Whole plantChloroform6.36 (3D7) and 5.24 (K1)[69]
Globba malaccensis (Ridl.)RhizomesEthanol1.50 (K1)[70]
Sanchus arvensis L.LeavesEthyl acetate2.9 (3D7)[71]
Terminalia arjuna (Roxb. Ex DC.) (Wight and Arn.)FruitsAqueous4.05 (K1)[72]
Terminalia bentzoe (L.) L.f.LeavesMethanol6.06 (Dd2)[73]
Bridelia atroviridis Müll. Arg.BarkHydroethanolic8.08 (Dd2)[74]
Annickia affinis (Exell) (Versteegh and Sosef)LeavesAqueous1.49 (3D7)[75]
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

Ribeiro, G.d.J.G.; Rei Yan, S.L.; Palmisano, G.; Wrenger, C. Plant Extracts as a Source of Natural Products with Potential Antimalarial Effects: An Update from 2018 to 2022. Pharmaceutics 2023, 15, 1638. https://doi.org/10.3390/pharmaceutics15061638

AMA Style

Ribeiro GdJG, Rei Yan SL, Palmisano G, Wrenger C. Plant Extracts as a Source of Natural Products with Potential Antimalarial Effects: An Update from 2018 to 2022. Pharmaceutics. 2023; 15(6):1638. https://doi.org/10.3390/pharmaceutics15061638

Chicago/Turabian Style

Ribeiro, Giovane de Jesus Gomes, Sun Liu Rei Yan, Giuseppe Palmisano, and Carsten Wrenger. 2023. "Plant Extracts as a Source of Natural Products with Potential Antimalarial Effects: An Update from 2018 to 2022" Pharmaceutics 15, no. 6: 1638. https://doi.org/10.3390/pharmaceutics15061638

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