Next Article in Journal
Taxonomic Revision of the Catostemma Clade (Malvaceae/Bombacoideae/Adansonieae)
Previous Article in Journal
Mamba-YOLO-ML: A State-Space Model-Based Approach for Mulberry Leaf Disease Detection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phytochemistry, Ethnopharmacology, and Pharmacology of Lessertia frutescens (Cancer Bush): A Comprehensive Review

Chemistry Department, Cape Peninsula University of Technology, Bellville Campus, Symphony Road, Bellville 7535, South Africa
*
Author to whom correspondence should be addressed.
Plants 2025, 14(14), 2086; https://doi.org/10.3390/plants14142086
Submission received: 31 May 2025 / Revised: 27 June 2025 / Accepted: 4 July 2025 / Published: 8 July 2025

Abstract

Lessertia frutescens (L.) Goldblatt & J.C.Manning (synonym Sutherlandia frutescens), commonly known as cancer bush, is one of the most prominently used South African medicinal plants, with a rich history of traditional uses among indigenous communities. Its phytochemical profile showed different metabolites such as amino acids, fatty acids, sugars, flavonoid glycosides, cycloartenol glycosides, and oleanane-type saponins. Moreover, several research studies have highlighted the promising therapeutic effects of L. frutescens in combating various cancer cell lines. Additionally, the plant demonstrated potent immunomodulatory, antioxidant, anti-inflammatory, antidiabetic, neuroprotective, antistress, and antimicrobial activities. These research findings highlight L. frutescens as a promising candidate for the development of new or complementary therapies for a range of diseases and conditions. This review analyses the chemical and biological properties of L. frutescens based on 154 articles identified through SciFinder. Of these, 78 articles, including two patents, met the inclusion criteria and were reviewed. Studies focused on agriculture and horticulture were excluded as they fell outside the scope of this research.

1. Introduction

Lessertia frutescens (formerly known as Sutherlandia frutescens), an indigenous Southern African medicinal plant belonging to the Fabaceae family, is prominent in traditional medicine [1]. Commonly referred to as “cancer bush”, it ranks among the foremost medicinal plants in South African herbal pharmacopoeia [2]. It is widely distributed across the Eastern Cape, KwaZulu-Natal, Northern Cape, and Western Cape, where it plays a crucial role in ameliorating the body’s ability to combat diseases and ailments. Moreover, it aids in reducing mental and physical stress by helping the body to mobilise its physiological and immunological resources [1,3,4].
Despite its extremely bitter taste, the leaves and stems of L. frutescens have been extensively studied for their therapeutic properties. Traditionally, prepared as a medicinal tea, the plant has been used for treating internal cancers and as a cancer prophylactic [2]. It is well known as an adaptogenic tonic, with commercial tablets frequently used to counteract the muscle-wasting associated with HIV-AIDS and to stimulate appetite. It is considered safe for consumption, with only mild side effects such as sporadic dry mouth, dizziness, mild diuresis, and diarrhoea in cachectic patients [3,5]. Furthermore, the plant has been used as a drug support in the treatment of anorexia, cancer, influenza, HIV/AIDS, and tuberculosis [6].
Scientific validation of these traditional claims has been pursued through various studies. In 2002, several clinical trials were conducted to verify the assertions made by indigenous people regarding the safety, potency, and therapeutic uses of L. frutescens. Following a three-month clinical trial assessing the plant’s toxicity using vervet monkeys, it was discovered that ingestion of the plant extract at human-equivalent dosages showed no toxicity or adverse effects. Consequently, the South African Medical Research Council (MRC) affirmed the safety of L. frutescens decoctions, infusions, and tinctures for consumption [7,8].
In vitro and in vivo studies have further highlighted its therapeutic potential, particularly in mitigating metabolic and oxidative stress-related disorders. Moreover, the isolated bioactive constituents such as mucronulatol, D-pinitol, sutherlandioside B, α-linolenic acid, L-canavanine, and GABA have been identified as key contributors to its neuroprotective, antidiabetic, antistress, anti-TB, and anticancer activities [4,9,10,11,12,13].
Despite its widespread use and documented therapeutic benefits, critical gaps remain in the literature. Previous reviews have predominantly focused on the plant extract’s anticancer, anti-inflammatory, anti-HIV, and immunomodulatory properties, often overlooking challenges like the high therapeutic concentrations required for efficacy in certain treatments. Furthermore, the documentation on L. frutescens secondary metabolites and their pharmacological properties is incomplete, emphasising the need for a comprehensive evaluation.
This review seeks to bridge existing knowledge gaps by critically synthesising the available literature on Lessertia frutescens, with a focus on its phytochemical composition, ethnobotanical significance, and pharmacological properties. Specifically, it aims to document the plant’s phytochemical constituents, assess the experimental approaches employed in studying its biological activities, including details such as concentrations, cell lines, organisms, and techniques, and to highlight its pharmacological potential while identifying limitations and gaps in current research.

2. Methodology

2.1. Search Strategy and Data Selection

This review was conducted without restrictions on geographical scope or time frame, with the search for articles concluding in December 2024. A total of 154 articles on S. frutescens (130) and L. frutescens (24) were identified using SciFinder. Of these, 78 articles, including 2 patents, were selected based on predefined inclusion criteria. These criteria focused on studies related to the phytochemistry, ethnopharmacology, and pharmacology of the plant, irrespective of the experimental methods, tested concentrations, or types of extracts.
Articles primarily addressing agriculture, horticulture, or other unrelated fields (66 in total) were excluded as they fell outside the scope of this review. Each selected article underwent a meticulous evaluation to ensure relevance and quality. References cited in the primary sources were further analysed to identify additional studies that aligned with the inclusion criteria.

2.2. Data Extraction

The data extraction process was designed to collect and organise detailed and relevant information aligned with the review’s objectives. Key data points included the phytochemical composition of the plant, as well as the therapeutic potentials of its extracts, fractions, and isolated compounds. Experimental approaches, including in vitro, in vivo, and clinical trials, were carefully documented. Data from these studies were categorised and tabulated to distinguish findings from in vitro, in vivo, and clinical trials, respectively.
Treatment protocols were meticulously detailed, encompassing doses or concentrations, treatment duration, targeted pharmacological activities, and the techniques used to evaluate these activities. Specific data points for the in vitro studies included the cell lines used, while the in vivo studies recorded the animal models and the conditions to which they were subjected. For clinical studies, data were gathered on the number of patients involved and their health status, specifying whether they were healthy or affected by a particular disease. Mechanisms of action for bioactive compounds and extracts were noted wherever reported. All the extracted data were carefully tabulated to ensure consistency and avoid duplication.

3. Taxonomy, Nomenclature, and Distribution

The Lessertia genus, belonging to the Fabaceae family, comprises 62 accepted species [14]. Some of these species were formerly classified under the Sutherlandia genus, such as Lessertia frutescens, previously known as Sutherlandia frutescens [14,15]. Prior to the reclassification of the Sutherlandia species, 35 species within the Lessertia genus were endemic to South Africa [16].
L. frutescens stands out as one of the most extensively studied and utilised medicinal plants in South Africa. It is known by names such as umnwele in Xhosa, blaasbossie in Afrikaans, Insiswa in Zulu, and Musa-Pelo in Sotho [17]. It is also associated with around twenty-five common names in languages like Afrikaans, Zulu, Tswana, and Sotho, which often reflect aspects of its characteristics, including its seedpods (blaasbossie and blaas-ertjie), flower colour or shape (kalkoenbos, hoenderbelletjie, and eendjie), appearance (unwele), taste (bitterbos), or medical uses (kankerbos, insiswa, phetola, and lerumo lamadi) [18].

4. Botanical Description

L. frutescens (Figure 1) is a perennial non-climbing shrub, typically reaching heights between 0.2 and 2.5 m [19]. Its leaves are greyish green, pinnately compound, with each leaflet measuring 4–10 mm. These leaflets vary from elliptic to narrowly oblong or ovate oblong, with the adaxial leaflets’ surface ranging from glabrous to sericeous depending on the plant’s cultivation region [4]. The prostate to erect stems is either sparsely pubescent or glabrous with many leaves in terminal racemes [4,20]. Its orange-red butterfly-shaped flowers (35 mm long) appear in short clusters within the leaf axils at branch tips from September to December [21]. After flowering, the plant produces inflated bladder-like pod fruits that contain black seeds [19,22].

5. Ethnomedicinal Uses

In South Africa, L. frutescens has been a staple in traditional medicine, utilised by healers, herbalists, diviners, and local people to treat various ailments and diseases. Despite its bitterness, it has gained popularity as a medicinal tea due to its liquorice aftertaste [15]. Since 1895, the Khoisan and Cape Dutch have known this flowering shrub as a cancer bush due to its potency against internal cancers [4,24,25]. Conversely, the Nama and Khoi-San communities traditionally use decoctions of the plant to treat fevers and wounds [25]. Historically, Zulu warriors would consume a concoction of the plant to induce relaxation following battle. In contrast, the widows of the deceased warriors used it as an antidepressant to help them navigate through their grief. In Van Wyk and Albrecht’s [4] review on the ethnobotany of L. frutescens, it was revealed that decoctions or infusions of the leaves were used in the treatment of diarrhoea, urinary tract infection, rheumatism, inflammation, intestinal pain, haemorrhoids, eye diseases, chickenpox, and skin disorders (Figure 2). Moreover, decoctions of L. frutescens have been used to treat various diseases and ailments such as asthma, chronic bronchitis, colds, coughs, convulsion, diabetes mellitus, epilepsy, gastric, gout, heart failure, heartburn, hypertension, kidney and liver infections, menopausal symptoms, osteoarthritis, pains, peptic ulceration, rheumatoid arthritis, reflux oesophagitis, varicose veins, and stress-related conditions linked to the endocrine system [6,8,26,27]. The plant serves as a tonic that cleanses the blood, stimulates appetite, and aids digestion [4,18,28].

6. Phytochemistry

The phytochemical studies conducted on L. frutescens have revealed that the leaves contain a high concentration of free amino acids such as L-asparagine (1.6–35 mg/g), (1); proline (0.7–7.5 mg/g), (2); and L-arginine (0.5–6.7 mg/g), (3) [4]. Additionally, the essential omega-3 fatty acid α-linolenic acid (4) was isolated from the dichloromethane–methanol (1:1) extract of the aerial part of the plant [11]. For the first time, Moshe [29] isolated L-canavanine (5), a non-protein amino acid usually found in the seed, from the leaves of L. frutescens. Furthermore, γ-aminobutyric acid (GABA), (6) was identified as another non-protein-free amino acid in the leaves. The cyclitol D-pinitol (7), known for its antidiabetic activity, was also isolated from the plant leaves. In addition to these compounds, researchers Fu et al. [30] isolated and identified four flavonoid glycosides, named sutherlandins A-D (811) (Figure 3).
Fu et al. [31,32,33] reported the isolation of 8 cycloartane glycosides given the trivial name of sutherlandiosides A-H (1421) (Figure 4). Recently, the oleanan-type saponin 3-O-[α-L-rhamnopyranosyl-(1-3)-β-D-glucurono pyranosyl]-22-epi-soyasapogenol B-22-O-β-D-glucopyranoside (31), along with seven cycloartane glycoside compounds named sutherlandiosides E-K (2227) were isolated [34]. However, some of these trivial names, specifically sutherlandiosides E-H (2225), have already been assigned by Fu [33] to another four cycloartane triterpenoid diglycosides (1821). Moreover, the cycloartane glycoside sutherlandioside I reported as new by Tchegnitegni et al. [34] had already been identified as sutherlandioside G (18) by Fu et al. [32]. The IUPAC names of the sutherlandiosides E-H isolated by Fu [33] and Tchegnitegni et al. [34] are tabulated in Table 1 to highlight the duplication in sutherlandiosides naming. Recently, Ndjoubi et al. [13] isolated two cycloartane glycosides, namely lessertiosides A (29) and B (30), as well as the flavonoids 8-methoxyvestitol (12) and mucronulatol (13).
In 2019, Gonyela et al. [35] reported the isolation of cycloartenol (28) from L. frustecens leaves. However, D-pinitol, L-canavanine, and sutherlandioside B are identified as the major components in the plant [36]. Apart from these secondary metabolites, L. frutescens is known to biosynthesise tannins [9,37]. As mentioned above, the chemistry of triterpenoids of this plant is unique, especially the oxygenation pattern of rings A and C. Compounds 20 and 21 have unique rearrangements in their cycloartenol glycoside structures, featuring a rearranged five- and seven-membered A/B-ring system. This discovery marked the first observation of the hexadecahydro-1-H-indeno [5,4-f] azulene ring system in nature [32]. Interestingly, compounds 16, 19, and 27 have a unique oxygenation pattern in rings A and C with two carbonyls at C-1 and C-11, and 3α-OH, while compounds 23 and 24 have 1α, 3α-diOH in addition to C=O in C-11. Another feature of the isolated triterpenoids is the configuration of the 3-OH, which is assigned to the uncommon α-position in most of the isolated compounds, except 20, which may need further revision.

7. Ethnopharmacological and Pharmacological Properties of L. frutescens Extracts and Compounds

L. frutescens, a medicinal plant native to Southern Africa, boasts diverse ethnomedicinal applications. In South Africa, the Khoisan and Cape Dutch people have historically used this perennial shrub for treating internal cancers, wounds, inflammation, stomach pains, diabetes, HIV/AIDS, and infections [4]. While the pharmacology and ethnomedicinal properties of L. frutescens (Table 2 and Table 3) have not been ascribed to a specific bioactive compound, it is believed that the synergy among the plant bioactive compounds contributes to its complex mechanism of action. L-canavanine, D-pinitol, and GABA are reported as the most bioactive elements within the plant.
The following sections will discuss a more detailed exploration of the biological activity of different organic extracts and bioactive compounds.

7.1. Cancer

The ethanolic extract of L. frutescens has demonstrated a significant cytotoxic effect on normal T-lymphocytes, particularly at a concentration of 2.5 mg/mL. After 24 h, the extract induced necrosis in 95% of cells, depleted ATP levels by 76%, and inhibited caspase 3/7 activity by 11%. In contrast, the deionised water extract at the same concentration caused milder effects, with necrosis at 26%, ATP levels at 91%, and caspase 3/7 inhibition at 15%. Both extracts exhibited time-dependent effects over 48 h, with the ethanolic extract showing more potent inhibition of cell growth through necrosis, ATP depletion, and reduced caspase activity. DNA fragmentation observed after 48 h confirmed the potential toxicity of the extracts, although the water extract appeared relatively safer [57].
Ethanolic extracts of L. frutescens have also shown anticancer activity. Tai et al. [9] reported that the ethanolic extract inhibited the proliferation of cancer cell lines, including Jurkat, MDA-MB-468 (malignant breast cancer), HL-60 (human leukaemia), and MCF-7 (breast cancer) with IC50 values of 0.91 mg/mL (1/150 dilutions), 0.68 mg/mL (1/200 dilutions), 0.68 mg/mL (1/200 dilutions), and 0.55 mg/mL (1/250 dilutions), respectively. The active compound L-canavanine, a non-proteinogenic amino acid, was implicated in the antiproliferative effects by inhibiting enzyme function and inducing protein misfolding [65]. Interestingly, L-arginine at 1 mM mitigated the antiproliferative effects of 2 mM L-canavanine in MCF-7 cells, suggesting a potential pathway to modulate toxicity. Further studies by Stander et al. [40] showed that a 70% ethanolic extract of L. frutescens (1.5 mg/mL) inhibited MCF-7 cell proliferation and induced apoptosis within 72 h. An aqueous extract at 10 mg/mL reduced cell growth by 26% in MCF-7 cells and 49% in MCF-12A cells. In MCF-7 cells, pronounced apoptotic changes, such as chromatin condensation and apoptotic bodies, were observed. Flow cytometry revealed a heightened sub-G1 apoptotic fraction and S-phase arrest. Transmission electron microscopy suggested that these effects were driven by autophagic and apoptotic processes, likely induced by L-canavanine’s protein misfolding response [54]. In contrast, Steenkamp and Gouws [66] reported that an aqueous extract (50 µg/mL) exhibited minimal cytotoxicity against MCF-7, DU-145 (prostate cancer), MDA-MB-231, and MCF-12A cells, suggesting that concentrations ≤ 50 µg/mL, the plant does not exhibit antiproliferative properties (Figure 5).
Methanolic extracts of L. frutescens have demonstrated cytotoxic effects against prostate cancer cell lines PC3, LNCaP, and TRAMP-C2, with IC50 values of 167, 200, and 100 µg/mL, respectively. These effects were independent of androgen receptor signalling and involved suppression of Gli/Hh signalling, as evidenced by reduced Gli1 and Ptch1 gene expression, which plays a role in prostate cancer tumorigenesis [10]. Similarly, ethanolic extracts have been shown to downregulate PI3K/Akt signalling, reduce FKHR phosphorylation, and activate mitochondrial apoptotic pathways in Caco-2 colon cancer cells, promoting apoptosis [39]. The aqueous extract at 2.63 mg/mL further induced cytotoxicity in LS180 colorectal cancer cells, depleting soluble protein content, intracellular ATP, and extracellular adenylate kinase within 24 h [41].
In melanoma and cervical cancer models, ethanolic extracts reduced the viability of melanoma cells (A-375 and Colo-800) by 62% and 43%, respectively, after 72 h at 0.625 mg/mL. It showed even greater efficacy against human dermal fibroblast cells (HDFα), where viability decreased by 81% at 0.3 mg/mL after 72 h [36]. Meanwhile, it was reported that the aqueous extract at 3.5 mg/mL induced apoptosis and cytotoxicity in Chinese hamster ovary (CHO) and cervical neoplastic cells [56]. Studies on oesophageal cancer (SNO cells) highlighted geographical variations in extract efficacy. Extracts from Colesberg induced apoptosis through caspase 3/7 activation, while extracts from Platvei triggered cytochrome c release, highlighting the influence of geographical variations on the phytochemical composition and biological activity of the plant [38].
Phulukdaree et al. [56] reported that L. frutescens aqueous extract (6 mg/mL) significantly reduced intracellular glutathione levels, increased lipid peroxidation, and induced mitochondrial membrane depolarisation (in 80% of the treated cells) in MDBK and LLC-PK1 cells. At higher concentrations (12 and 24 mg/mL), the extract increased oxidative stress, disrupted mitochondrial integrity, and promoted apoptosis. These findings suggest that L. frutescens aqueous extract has dose-dependent cytotoxic effects on MDBK and LLC-PK1 cells, mediated primarily through the induction of oxidative stress and mitochondrial damage (Figure 6).

7.2. COVID-19

A molecular docking investigation revealed that L-canavanine is a promising inhibitor of the SARS-CoV-2 3CLPro, showing favourable binding modes and strong interactions in the active site of 3CLPro [68]. Moreover, Akindele et al. [69] also reported that apart from its antiviral, anti-inflammatory, and immunomodulatory properties, the plant also possesses COVID-19 symptom-relieving activity.

7.3. Antioxidant

The ethanolic extract of L. frutescens demonstrated significant hydroxyl radical scavenging in the TEAC assay but failed to modulate LPS-induced NO production in RAW 264.7 cells across various concentrations ranging from 0.068 to 0.68 mg/mL. In contrast, L-canavanine (0.5 mM) and D-pinitol (10 mM) significantly inhibited LPS-induced NO secretion. Given L-canavanine’s role as a selective inhibitor of iNOS, the absence of inhibitory activity by L. frutescens may be concentration-dependent [9]. Similarly, Fernandes et al. [42] reported that a hot aqueous extract of L. frutescens at a concentration of 10 µg/mL reduced the luminol- and lucigenin-enhanced chemiluminescence response in FMLP-stimulated neutrophils. In the hydrogen peroxide/horseradish peroxidase-mediated chemiluminescence, it scavenged neutrophil-derived oxidants at 2.5 µg/mL. Furthermore, at a concentration of 0.62 µg/mL, it inhibited horseradish peroxidase/hydrogen peroxide-induced chemiluminescence [42]. Moreover, the antioxidant efficiency of L. frutescens varied significantly depending on the extraction solvent [44]. This variation is primarily attributed to the solvent’s impact on the composition of phenolics and flavonoids in the extract. Solvents with higher polarity yielded extracts with greater total phenolic and flavonoid content, resulting in greater reducing power and radical scavenging activity [44,70]. The freeze-dried hot water extract of L. frutescens (500 µg/mL) demonstrated protective effects against tert-butyl hydroperoxide (t-BHP)-induced oxidative stress in CHO, human hepatoma (HepaRG), and human pulmonary alveolar carcinoma (A549) cells by effectively scavenging ROS and preserving intracellular glutathione (GSH/GSSG) levels. At a 1 mg/mL concentration, the extract exhibited potent scavenging activity, effectively neutralising hydroxyl radicals, followed by superoxide radicals and hydrogen peroxide [44].

7.4. Immune Modulation and Inflammation

Na et al. [71] reported that the methanolic extract (10, 5, and 1 µg/mL) of L. frutescens inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced cyclooxygenase-2 (COX-2) expression in human breast epithelial (MCF10A) cells by suppressing the DNA-binding activity of nuclear factor kappa-light chain enhancer of activated B cells (NF-κB) induced by TPA (10 nM). This inhibition of TPA-induced COX-2 expression, achieved through suppressing NF-κB DNA binding, may be responsible for the plant’s chemopreventive activity [71]. The aqueous extract also partially reduced tumour necrosis factor-alpha (TNF-α) induced chemokine CCL5 expression in NRK-52E cells [46]. The transcriptome analysis has provided valuable insights into the role of L. frutescens in modulating the immune system. The crude polysaccharide-enriched fraction of L. frutescens aqueous extract influenced gene expression in activated murine macrophage cell lines (RAW 264.7), resulting in the differential expression of 547 genes [20,51]. This fraction also exhibited immuno-stimulatory activity by activating macrophages via TLR4 receptors and the NF-κB signalling pathway [51]. Additionally, the ethanolic extract-enriched polysaccharides fraction reduced the production of nitric oxide (NO) and reactive oxygen species (ROS), as well as inhibited the phosphorylated extracellular signal-regulated kinase ½ (p-ERK1/2), signal transducer, and activator of transcription 1-α (STAT1-α) and NF-κB induced by lipopolysaccharides (LPS) and interferon-gamma (IFNγ) [45]. Furthermore, the ethanolic and aqueous extracts were found to significantly inhibit GM-CSF, G-CSF, IL-1α, IL-6, TNF-α, iNOS, NO, ROS, COX-2, and CD86 (Figure 7). The ethanolic extract also modified the M1 and M2 macrophage phenotypes’ expressions by enhancing the M2 phenotype and downregulating the M1 phenotype [50]. These findings corroborate the results of Lei et al. [45] regarding the plant anti-inflammatory macrophage markers.
L. frutescens was also shown to negatively regulate the NF-κB signalling pathway by suppressing NF-κB nuclear translocation following LPS induction. This further substantiates the plant’s ability to inhibit NF-κB activation through the attenuation of NF-κB p65 subunit phosphorylation on the Ser 536 residue, which is essential for both NF-κB nuclear transcriptional and translocation activity [50]. Likewise, Kirsten [72] stated that the plant extract regulated the expression of IL-6, IL-10 and IFN-γ in phytohaemagglutinin (PHA) and LPS. However, it was reported that these immune modulation effects are donor-dependent [72]. Additionally, little effect of the ethanol extract on the stimulation of TNF-α and IL-8 by phorbol myristoyl acetate was observed [43,73]. Jiang et al. [43] further demonstrated that the ethanol extract could mitigate N-methyl-D-aspartic acid (NMDA) induced neuronal oxidative responses and reduce ROS and NO production induced by LPS and IFN-γ in microglial cells (BV-2 and HAPI).
Moreover, the ability of the ethanolic extract to inhibit IFN-γ-induced p-ERK1/2 pathway explains the extract’s potential in preventing or treating inflammatory infections, including HIV-associated neurocognitive disorders. L. frutescens shoot aqueous extract inhibited fresh egg albumin-induced acute inflammation, triggering hypoglycemia in rats [3,69]. Its efficacy as an antidiabetic and anti-inflammatory herbal remedy can be attributed to its inhibitory effects on cytokines and apoptosis [64,74,75]. Furthermore, it also found that the aqueous extract inhibited the gene expression of CYP3A4 and CYP2D6 enzymes instead of inducing them [41].

7.5. Nephrotoxicity

The sugar D-pinitol improved histopathological alterations in cisplatin-induced nephrotoxicity in mice because of its antiapoptotic, antioxidant, and anti-inflammatory properties [64]. Likewise, the reduction in histopathological and biochemical alterations, as well as decreasing levels of cytokines (TNF-α, IL-6, and IL-1β) and oxidative stress in cisplatin-induced nephrotoxicity, ameliorate the nephrotoxic reaction of cisplatin in D-pinitol-treated mice [64].

7.6. Antimicrobial

The IC50 of L. frutescens hexane extract against Enterococcus faecali, Escherichia coli, and Staphylococcus aureus were found to be 2.50, 1.25, and 0.31 mg/mL, respectively [60]. Conversely, the dichloromethane–methanol (1:1) extract displayed a good inhibition against the shikimate kinase enzyme, an important drug target for Mycobacterium tuberculosis, with an IC50 of 0.1 μg/mL, whereas the aqueous and ethanolic extracts had IC50 of 5.1 and 1.7 μg/mL, respectively [11]. The efficiency of the dichloromethane–methanol (1:1) extract as shikimate kinase inhibitor was attributed to the essential omega-3 fatty acid α-linolenic acid, which is known for its antimicrobial activity against Staphylococcus aureus, Bacillus subtilis, Helicobacter pylori, Rhizoctonia solani, Crinipellis perniciosa, and hepatitis C virus [11,75,76,77,78]. α-linolenic acid was found to possess antitubercular activity by inhibiting the shikimate kinase enzyme with an IC50 of 3.7 μg/mL [11].
The ethyl acetate and 50% methanolic extracts were diluted to 5%, 10%, and 20% (w/w) with DMSO and tested for their mutagenic and antimutagenic properties against Salmonella typhimurium strains TA97a, TA98, TA100, and TA102. After investigations, it was observed that the ethyl acetate extract significantly exhibited antimutagenic effects against TA97a, TA98, TA100, and TA102 [58]. On the other hand, the methanolic extract showed pro-mutagenic and antimutagenic potential in the presence of the S9 in TA98 with 2-acetamidofluorene and TA100 with aflatoxin B1. L-arginine, GABA, and D-pinitol exhibited antimutagenic activity against all four strains, whilst L-canavanine displayed a co-mutagenic effect in the absence of S9 in TA97 with 9-aminoacridine. Thus, the pro-mutagenic activity of the methanol extract cannot be ascribed to L-canavanine [58]. The ethyl acetate extract, having a higher antimutagenic potential and total phenolic content than the methanolic extract, explains the correlation between the antioxidant and antimutagenic activities of the plant.

7.7. HIV/AIDS

The aqueous extract (200 µg/mL) of the leaves was reported to inhibit the HIV-1 reverse transcriptase (RT) enzyme (Figure 8) by ≥50% [47]. However, when tested with 0.2% (w/v) bovine serum albumin (BSA) to neutralise tannin effects, the inhibitory activity was reduced, indicating that tannins contributed significantly to the inhibition. Despite this, the extract retained approximately 30% of its activity. On the other hand, the dichloromethane extract exhibited limited activity against the HIV-II protease enzyme but significantly inhibited α- and β-glucosidase enzymes [47].
An ethanolic extract concentration equivalent to its calculated IC50 (7.5 mg/mL) was administered to normal human lymphocytes for 3, 6, and 12 h. At the 12 h mark, the extract induced apoptosis in total lymphocytes, with a stronger effect on CD4+ subpopulations. This was supported by increased caspase-3/7 activity, phosphatidylserine (PS) translocation, and reduced ATP levels [79]. Additionally, after 12 h, the extract doubled the number of lymphocytes expressing the CD69 activation marker, leading to activation-induced cell death. These findings contradicted earlier clinical suggestions that the extract might be useful in treating HIV/AIDS [79].
D-pinitol and GABA have been proposed to alleviate wasting conditions in cancer and HIV/AIDS patients by inhibiting inflammatory cytokines TNF-α and IL-1β, thus enhancing glucose availability for cell metabolism [79,80,81]. Conversely, L-canavanine demonstrated antiviral properties against HIV and influenza by disrupting viral protein synthesis and function [79,82]. Chronic oral administration of L. frutescens extract (12 mg/kg for 5 days) induced intestinal and hepatic CYP3A2 expression in rats, altering the pharmacokinetics of the antiretroviral drug nevirapine and increasing CYP3A4 activity in LS180 cells [62]. This suggests a potential drug–herb interaction when the nevirapine is co-administered with L. frutescens. D-pinitol and the aqueous extract reduced atazanavir accumulation in Caco-2 cells at 10 mg/mL, potentially lowering its bioavailability, while a triterpenoid glycoside-enriched fraction enhanced atazanavir accumulation and absorption [52]. Additionally, the methanolic and aqueous extracts of L. frutescens inhibited atazanavir metabolism in human liver microsomes, indicating a potential impact on the drug’s clinical metabolism and absorption [52].
Fasinu et al. [53] also demonstrated the anti-HIV activity of L. frutescens against various cytochrome P450 isozymes. These include CYP1A2-mediated phenacetin demethylation, CYP2A6-mediated coumarin 7-hydroxylation, CYP2B6-mediated bupropion hydroxylation, CYP2C8-mediated paclitaxel 6α-hydroxylation, CYP2C9-mediated diclofenac 4′-hydroxylation, CYP2C19-mediated S-mephenytoin 4′-hydroxylation, CYP3A4/5-mediated midazolam 1′-hydroxylation, and CYP3A4/5-mediated testosterone 6β-hydroxylation in pooled human liver microsomes (Figure 9) with IC50 values of 41, 160, 20, 22.4, 23, 35.9, 17.5, and 28.3 μg/mL, respectively [53]. The studied extract induced time-dependent (irreversible) inhibition of CYP3A4/5 with an inhibition constant (Ki) of 296 μg/mL and a maximal rate of enzyme inactivation (Kinact) of 0.063 min−1 [53]. The authors also indicated that the plant inhibited the human ATP-binding cassette transporters P-gp as well as the organic anion transport polypeptide OATP1B1 and OATP1B3 with IC50 values of 324.8, 10.4, and 6.6 μg/mL, respectively. This inhibition also led to a 40% reduction in the clearance of midazolam metabolites in hepatocytes. However, no activity was observed when treating the efflux transporter BRCP (breast cancer resistance protein) as well as the enzymes CYP2D6 and CYP2E1 with L. frutescens [53].
Despite some therapeutic potential, L. frutescens also raised safety concerns. Africa and Smith [83] found that the plant significantly reduced IL-1β secretion but increased monocyte chemoattractant protein-1 (MCP-1) levels, leading to greater infiltration of CD14+ monocytes across the blood–brain barrier (Figure 10). This exacerbated HIV-associated neuroinflammation, prompting warnings against its use by HIV patients at any stage of infection [83,84].

7.8. Neuroprotection

Pre-treatment of 1-methyl-4-phenylpyridinium (MPP+) induced toxicity in SH-SY5Y neuroblastoma cells, with the plant aqueous extract resulting in the protection of the cells from the MPP+ induced toxicity and loss of MPP via the regulation of ROS, thus hinting at the extract’s neuroprotective effect and its potential as an anti-Parkinson agent [49]. In a study by Ndjoubi et al. [13], several natural compounds, including 8-methoxyvestitol; mucronulatol; proline; D-pinitol; sutherlandin C; sutherlandiosides B, D, K; and 7S,24S,25-trihydroxy-9,10R-seco-9,19-cyclolanost-2(3),9(11)-diene-25-O-β-D-glucopyranoside, demonstrated significant neuroprotective effects through their antiapoptotic activity. The compounds were evaluated for their antiapoptotic potency, with sutherlandioside B, mucronulatol, proline, and D-pinitol significantly restoring ATP levels from 51% (MPP+-treated) to 73, 75, 74, and 75%, respectively, while inducing caspase 3/7 activity from 5 fold to 1.5–2.8 fold relative to controls, with mucronulatol exhibiting the most potent antiapoptotic effect (1.5-fold) [13].

7.9. Diabetes

Studies have shown the effectiveness of L. frutescens extracts in managing diabetes through various mechanisms. Oral administration of the shoot aqueous extract strongly inhibited streptozotocin-induced hyperglycemia in mice at concentrations ranging from 50 to 800 mg/kg [3]. Additionally, the aqueous leaf extract showed promise as a type 2 antidiabetic drug by significantly increasing glucose uptake into muscle and adipose tissue while significantly decreasing intestinal glucose uptake (after 1 h). This indicates the extract’s potential to normalise insulin levels and glucose uptake in peripheral tissues and suppress intestinal glucose uptake without causing weight gain [20,61]. Studies on rats fed on a high-fat diet showed that the plant extracts prevent the development of insulin resistance by reducing plasma-free fatty acid levels [85]. It was also observed that rats on a high-fat diet exhibited a twelve-fold reduction in plasma-free fatty acid levels compared to those on a normal diet [85]. Moreover, Bates et al. [86] stated that D-pinitol acted similarly to insulin by lowering blood sugar levels and augmenting glucose uptake for cellular metabolism. This resulted in its capacity to regulate cellular energy by boosting energy levels and reducing fatigue. In 2013, it was discovered that the aqueous extract could prevent insulin resistance in hepatocytes [48].

7.10. Stress

The warm water extract of L. frutescens leaves was revealed to efficiently reduce the corticosterone response to chronic stress in Wistar rats [59]. This finding confirmed the traditional use of the plant in treating ailments associated with high levels of glucocorticoids. Investigations on the aqueous and methanolic extracts revealed these extracts inhibit progesterone (PROG) binding to CYP17A1 and CYP21A2 without affecting 3β-HSD2 [59]. The methanolic extract containing sutherlandioside B (SUB) as its major component significantly inhibited pregnenolone (PREG)and PROG conversion by CYP17A1. Interestingly, at lower concentrations, the extract could considerably affect the catalytic activity of CYP17A1 only by PROG conversion [12]. The absence of an inhibitory effect on PREG metabolism suggests that the plant’s bioactive compounds may bind to a site in the active pocket other than the one occupied by PREG [59]. Changes observed in the inhibition of PROG and PREG metabolism and substrate binding imply that the extract’s bioactive components probably act synergistically and interfere with the electron transport chain to inhibit CYP17A1 and CYP21A2 enzymes [59]. Furthermore, SUB was reported to inhibit CYP17A1 towards PREG and PROG as well as 3β-HSD2, signifying that SUB could disrupt steroidogenesis at the branch point [12]. In human H295R adrenal cells, the extract inhibited CYP11B1 by considerably reducing cortisol (CORT) and 11-hydroxy androstenedione (11-OHA4) levels, explaining the plant’s antistress, anti-anxiety, and anti-hypertensive properties [12]. Moreover, the methanol extract and SUB acted as selective glucocorticoid receptor agonists (SEGRAs) by not showing any transactivation ability on glucocorticoid response element-driven gene expression. SUB and the studied plant extract also suppressed NF-κB -driven gene expression while being unable to activate mineralocorticoid receptor (MR) mediated gene transcription, although both antagonised the effects of aldosterone via MR [12].
The non-protein free amino acid GABA exhibits anti-neurotransmitter properties, which partly explain the use of L. frutescens for stress and anxiety disorders [36]. The anti-anxiety activity of GABA has been linked to its ability to reduce glucocorticoid production [80].

7.11. Toxicology

The traditional dosage involves daily infusions or decoctions of 2.5–5 g of dried material. The highest recorded dose, a decoction of 5 g of leaves, stems, and pods taken twice daily over six years, resulted in no adverse effects [4]. Furthermore, studies on the intraperitoneal administration of graded aqueous extracts of L. frutescens in fasted Balb C albino mice (20–25 g) established the lethal dose (LD50) at 1280 ± 71 mg/kg, suggesting that the crude extracts are likely to be relatively safe in mammals [3]. A study on determining the toxicity of the aqueous and ethanolic leaf extracts on zebrafish embryos, focusing on their hatching rates and larval mortality at concentrations ranging from 5 to 300 µg/mL, exhibited lethal concentration (LC50) values of 297.57 µg/mL (aqueous) and 40.54 µg/mL (ethanol), reaffirming the claim that the water extract is less toxic than the ethanol extract [84]. However, further study on how the plant may interact with other drugs and diseases is essential to avoid fatal or detrimental side effects.
For commercial preparations, a recommended dose of 300 mg of dried leaves twice daily (600 mg/day) is advised, with the caution that it should be avoided during pregnancy and lactation. This conservative dosage was used for a safety study in vervet monkeys, where doses of 0, 9, 27, and 81 mg/kg body weight correspond to 0, 1, 3, and 9 times the recommended human dose. These doses were administered as part of a standard diet for three months, and the study showed no clinical side effects across 15 haematological, 21 clinical biochemical, 6 physiological, and many behavioural variables, providing strong and reassuring evidence of the safety of L. frutescens at recommended human doses [4].

7.12. Clinical Trials

Grandi et al. [87] conducted a study with 16 cancer patients (11 men and 5 women) to evaluate the effect of 600 mg/day of aqueous L. frutescens extract (Figure 11). They found that the extract significantly decreased fatigue in cancer patients, with no other major adverse effects reported (Table 4, Figure 10). In Johnson et al. [5], a randomised, double-blind, placebo-controlled trial involving 25 healthy adults examined the effects of 800 mg/day of L. frutescens leaf powder capsules. The study concluded that the powder was well tolerated over three months, with no significant adverse events observed, and there was a noted improvement in appetite in the treatment group [5]. In 2002, the South African Ministry of Health recommended the aqueous extract as a drug support in the treatment of HIV/AIDS [8] as it decreased viral loads and improved CD4 counts [88]. However, preclinical studies performed in 2011 have indicated that using L. frutescens extract alongside antiretroviral drugs or CYP3A4 substrates may cause harmful drug–herb interactions, treatment failure, and the development of viral resistance [8,62]. Wilson et al. [89] performed a study on 107 participants, dividing them into two groups: one received 2400 mg/day of L. frutescens leaf powder (1200 mg twice daily), and the other received a placebo. The results showed that L. frutescens did not alter the viral load or CD4 T-lymphocyte count, but the treatment group had a higher burden, primarily due to two tuberculosis cases in patients on isoniazid preventive therapy (IPT). While no other safety concerns related to L. frutescens consumption were detected, the study indicates the need for further investigation into the potential interaction between L. frutescens and IPT [89].

8. Discussion

L. frutescens has been extensively studied for its anticancer, anti-inflammatory, immune booster and anti-HIV properties, with traditional usage suggesting minimal side effects. However, research studies highlight significant complexities and limitations in its application, particularly in anticancer and anti-HIV therapies, while revealing promise in other areas such as immune booster, metabolic, oxidative, and microbial conditions.
The cytotoxic effects of L. frutescens extracts have been demonstrated in various cancer cell lines. These effects are primarily mediated through mechanisms such as PI3K/Akt inhibition, oxidative stress, mitochondrial dysfunction, apoptosis, caspase activation, and suppression of the Gli/Hh signalling pathway. Despite these promising results, the therapeutic concentrations required (0.3–10 mg/mL) are significantly higher than those of standard chemo-therapeutic drugs like doxorubicin (IC50 = 0.68 ± 0.04 μg/mL for MCF-7) and paclitaxel (IC50 = 2.5 ng/mL for MCF-7 and 2.6 ng/mL for HeLa), raising concerns about the practical application of L. frutescens in cancer therapy [90,91]. Additionally, the lack of selectivity is a major limitation, as both cancer and normal cells (T-lymphocytes) exhibit toxicity at similar concentrations. This raises questions about the plant’s therapeutic index and clinical safety for cancer patients.
In HIV therapy, L. frutescens has shown inhibitory effects on HIV-1 RT, primarily due to its tannin content. However, tannins lack specificity, and their pharmacological efficacy is significantly lower than the established antiretroviral drugs [92]. For instance, tenofovir, a nucleotide analogue RT inhibitor, exhibits highly specific activity by competing with the natural substrate, deoxyadenosine 5′-triphosphate, for incorporation into viral DNA at the active site of reverse transcriptase, ultimately causing premature termination of the DNA chain during replication with IC50 values of 0.5–2.2 µM with a highly specific mechanism of action [93,94,95]. On the other hand, L. frutescens extracts require much higher concentrations (IC50 = 200 µM), limiting their practical utility.
Furthermore, the immunotoxicity of L. frutescens further complicates its potential use in HIV therapy. At an IC50 concentration of 7.5 mg/mL, the ethanolic extract induces apoptosis in lymphocytes and CD4+ cells by increasing caspase-3/7 activity, promoting phosphatidylserine translocation, and depleting ATP levels. This activation-induced cell death significantly undermines the preservation of the CD4+ T-cell population, a critical goal in HIV treatment [96]. The resulting loss of immune integrity heightens susceptibility to opportunistic infections, such as tuberculosis and fungal infections, presenting a substantial drawback for therapeutic application.
Its potential for drug–herb interactions poses significant challenges, as it may compromise the efficacy and bioavailability of antiretroviral drugs, creating risks for patients who require precise drug concentrations. Additionally, L. frutescens has been associated with exacerbating HIV-associated neuroinflammation, evidenced by increased MCP-1 levels and CD14+ monocyte infiltration across the blood–brain barrier. These effects could worsen HIV-associated neurocognitive impairment, a condition affecting 42.6% of patients [96,97]. Despite the higher concentrations required for anticancer and anti-HIV activity, L. frutescens has demonstrated more promising potential in other therapeutic areas. Studies highlight significant antidiabetic, antioxidant, neuroprotective, anti-inflammatory, and antimicrobial properties. Both aqueous and ethanolic extracts, along with bioactive compounds such as sutherlandioside B, mucronulatol, D-pinitol, and α-linolenic acid, exhibit potent biological activity at concentrations below 50 µg/mL (Table 2 and Table 3). For instance, D-pinitol is recognised for its insulin-sensitising effects, while α-linolenic acid, sutherlandioside D, and mucronulatol contribute to the plant’s anti-tubercular, antistress, and neuroprotective activities, respectively. Such properties underscore the plant’s potential in addressing metabolic and oxidative stress-related disorders.

9. Economic Importance of L. frutescens

In South Africa, the commercial value of L. frutescens lies in its renowned ethnopharmacological applications, particularly in treating internal cancers, HIV/AIDS, and diabetes. Additionally, it is prized for its immune-boosting and antioxidant properties and its use as a skincare product. Over the past decade, the plant has been sold in various forms, including capsules, tablets, teas, syrup, soap, cream, and raw materials products, as shown in Figure 12.
Moreover, the global trade of L. frutescens extends to regions such as North and South America, Western and Eastern Europe, Asia-Pacific, the Middle East, and Africa, with Africa and the Middle East holding a significant share of the global trade. Different South African companies are leading retailers and distributors of the various processed and semi-processed forms of L. frutescens [98].
Its popularity in traditional medicine has sparked significant scientific interest in understanding the pharmacokinetics and pharmacodynamics of L. frutescens crude extracts and identifying the specific metabolites responsible for its ethnobotanical properties and its ability to treat conditions related to oxidative stress. These investigations have prompted companies and researchers to patent their formulations and extraction methods for sale [99,100].

10. Conclusions

The phytochemical investigation on L. frutescens has highlighted its potential as a rich source of amino acids, flavonoid glycosides, and cycloartane triterpenes glycosides. The discovery of novel cycloartane glycosides and 9,10-seco-cycloartane-type diglycosides with an unprecedented 5/7/6/5 ring skeleton highlights their significance in natural product chemistry. Moreover, the oxygenation pattern of rings A and C of many of the isolated compounds is unique and limited to this plant.
Despite its promising bioactivity, the therapeutic efficiency of L. frutescens faces challenges, including its high effective concentrations, lack of selectivity, potential drug–herb interactions, and immunotoxicity. Most observed activities occur at concentrations exceeding the 50 µg/mL threshold, raising concerns about its clinical relevance for cancer and HIV treatment. Nonetheless, its potential in managing diabetes, neurodegenerative disorders, microbial infections, and oxidative-related conditions presents an opportunity for further exploration.

Funding

This research was funded by South African NRF, grant number 106055 under Ahmed A. Hussein.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Van Wyk, B.E.; Van Oudtshoorn, B.; Gericke, N. Medicinal Plants of South Africa; Briza Publications: Pretoria, South Africa, 1997; pp. 205–515. [Google Scholar]
  2. Mncwangi, N.; Viljoen, A.; Mulaudzi, N.; Fouche, G. Lessertia frutescens. In The South African Herbal Pharmacopoeia; Academic Press: Cambridge, MA, USA, 2023; pp. 321–344. [Google Scholar]
  3. Ojewole, J. Analgesic, antiinflammatory and hypoglycemic effects of Sutherlandia frutescens R. BR. (variety Incana E. MEY.) [fabaceae] shoot aqueous extract. Methods Find. Exp. Clin. Pharmacol. 2004, 26, 409–416. [Google Scholar] [CrossRef] [PubMed]
  4. Van Wyk, B.E.; Albrecht, C. A review of the taxonomy, ethnobotany, chemistry and pharmacology of Sutherlandia frutescens (Fabaceae). J. Ethnopharmacol. 2008, 119, 620–629. [Google Scholar] [CrossRef] [PubMed]
  5. Johnson, Q.; Syce, J.; Nell, H.; Rudeen, K.; Folk, W. A randomized double-blind placebo-controlled trial of Lessertia frutescens in Healthy Adults. PLoS Clin. Trial 2007, 2, e16. [Google Scholar] [CrossRef] [PubMed]
  6. Gericke, N.; Albrecht, C.F.; Van Wyk, B.; Mayeng, B.; Mutwa, C.; Hutchings, A.  Sutherlandia frutescens. Aust. J. Med. Herb. 2001, 13, 9–15. [Google Scholar]
  7. Morris, K. Treating HIV in South Africa—A tale of two systems. Lancet 2001, 357, 1190. [Google Scholar] [CrossRef]
  8. Mills, E.; Cooper, C.; Seely, D.; Kanfer, I. African herbal medicines in the treatment of HIV: Hyoxis and Sutherlandia frutescens: An overview of evidence and pharmacology. Nutr. J. 2005, 4, 19–24. [Google Scholar] [CrossRef]
  9. Tai, J.; Cheung, S.; Chan, E.; Hasman, D. In vitro culture studies of Sutherlandia frutescens on human tumor cell lines. J. Ethnopharmacol. 2004, 93, 9–19. [Google Scholar] [CrossRef]
  10. Lin, L.G.; Ung, C.O.L.; Feng, Z.L.; Huang, L.; Hu, H. Naturally occurring diterpenoid dimers: Source, biosynthesis, chemistry, and bioactivities. Planta Med. 2016, 82, 1309–1328. [Google Scholar] [CrossRef]
  11. Masoko, P.; Mabusa, I.H.; Howard, R.L. Isolation of α-linolenic acid from Sutherlandia frutescens and its inhibition of Mycobacterium tuberculosis shikimate kinase enzyme. BMC Complement. Altern. Med. 2016, 16, 366. [Google Scholar] [CrossRef]
  12. Sergeant, C.A.; Africander, D.; Swart, P.; Swart, A.C. Sutherlandia frutescens modulates adrenal hormone biosynthesis, acts as a selective glucocorticoid receptor agonist (SEGRA), and displays anti-mineralocorticoid properties. J. Ethnopharmacol. 2017, 202, 290–301. [Google Scholar] [CrossRef]
  13. Ndjoubi, K.O.; Omoruyi, S.I.; Luckay, R.C.; Hussein, A.A. Isolation of lessertiosides A and B and other metabolites from Lessertia frutescens and their neuroprotective activity. Plants 2024, 13, 3076. [Google Scholar] [CrossRef] [PubMed]
  14. Plants of the World Online (POWO). Lessertia frutescens (L.) Goldblatt & J.C. Manning. Available online: https://powo.science.kew.org/results?q=sutherlandia%20frutescens (accessed on 22 June 2023).
  15. Van Wyk, B.E.; Gorelik, B. The history and ethnobotany of Cape herbal teas. S. Afr. J. Bot. 2017, 110, 18–38. [Google Scholar] [CrossRef]
  16. Balkwill, M.J.; Balkwill, K. The genus Lessertia DC. (Fabaceae-Galegeae) in KwaZulu-Natal (South Africa). S. Afr. J. Bot. 1999, 65, 339–356. [Google Scholar] [CrossRef]
  17. Jackson, W.P.U. Origins and Meanings of Names of South African Plant Genera; U.C.T. Printing Department: Cape Town, South Africa, 1990. [Google Scholar]
  18. Aboyade, O.M.; Styger, G.; Gibson, D.; Hughes, G. Sutherlandia frutescens: The meeting of science and traditional knowledge. J. Altern. Complement. Med. 2013, 20, 71–76. [Google Scholar] [CrossRef]
  19. Schrire, B.D.; Andrews, S. Sutherlandia: Gansies or balloon peas. Plantsman 1992, 14, 65–69. [Google Scholar]
  20. PlantzAfrica. Sutherlandia frutescens Herba. Available online: http://www.plantzafrica.com/medmonographs/sutherlfrut.pdf (accessed on 3 February 2023).
  21. Adefuye, O.J. Anti-Diabetic and Phytochemical Analysis of Sutherlandia fruescens Extracts. Ph.D. Thesis, Nelson Mandela University, Port Elizabeth, South Africa, 2016. [Google Scholar]
  22. Daghman, M.I. Comparison of the Physicochemical Characteristics and Flavonoid Release Profiles of Sutherlandia frutescens Phytosomes Versus Liposomes. Master’s Thesis, University of Western Cape, Cape Town, South Africa, 2015. [Google Scholar]
  23. Zdeněk, H. BioLib.cz. Available online: https://www.biolib.cz/en/image/id325772/ (accessed on 15 May 2021).
  24. Drewes, S.E. Natural products research in South Africa: 1890–2010. S. Afr. J. Sci. 2012, 108, 1–8. [Google Scholar] [CrossRef]
  25. Street, R.A.; Prinsloo, G. Commercially important medicinal plants of South Africa: A review. J. Chem. 2012, 2013, 205048. [Google Scholar] [CrossRef]
  26. Prevoo, D.; Smith, C.; Swart, P.; Swart, A.C. The effect of Sutherlandia frutescens on steroidogenesis: Confirming indigenous wisdom. Endocr. Res. 2004, 30, 745–751. [Google Scholar] [CrossRef]
  27. Ojewole, J.A.O. Antinociceptive, anti-inflammatory, and antidiabetic properties of Hypoxis hemerocallidea Fisch. & C.A. Mey (Hypoxidaceae) corm [‘African Potato’] aqueous extracts in mice and rats. J. Ethnopharmacol. 2008, 103, 126–134. [Google Scholar]
  28. Thring, T.S.; Weitz, F.M. Medicinal plant use in the Bredasdorp/Elim region of the Southern Overberg in the Western Cape Province of South Africa. J. Ethnopharmacol. 2006, 103, 261–275. [Google Scholar] [CrossRef]
  29. Moshe, D.; Van der Bank, H.; Van der Bank, M.; Van Wyk, B.E. Lack of genetic differentiation between 19 populations from seven taxa of Sutherlandia. Tribe: Galegeae, Fabaceae. Biochem. Syst. Ecol. 1998, 26, 595–609. [Google Scholar] [CrossRef]
  30. Fu, X.; Li, X.C.; Wang, Y.H.; Avula, B.; Smillie, T.J.; Mabusela, W.; Syce, J.; Johnson, Q.; Folk, W.; Khan, I.A. Flavonol glycosides from the South African medicinal plant Sutherlandia frutescens. Planta Med. 2010, 76, 178–181. [Google Scholar] [CrossRef] [PubMed]
  31. Fu, X.; Li, X.C.; Smillie, T.J.; Carvalho, P.; Mabusela, W.; Syce, J.; Johnson, Q.; Folk, W.; Avery, M.A.; Khan, I.A. Cycloartane glycosides from Sutherlandia frutescens. J. Nat. Prod. 2008, 71, 1749–1753. [Google Scholar] [CrossRef]
  32. Fu, X.; Li, X.C.; Smillie, T.J.; Khan, I.A. Rearranged Cycloartanol Glycosides from Sutherlandia frutescens. Planta Med. 2010, 76, 47. [Google Scholar] [CrossRef]
  33. Fu, X. Phytochemical Studies on the Medicinal Plant Sutherlandia frutescens. Ph.D. Thesis, University of Mississippi, Oxford, MS, USA, 2012. [Google Scholar]
  34. Tchegnitegni, B.T.; Lerata, M.S.; Beukes, D.R.; Antunes, E.M. Sutherlandiosides E–K: Further cycloartane glycosides from Sutherlandia frutescens. Phytochem. Lett. 2024, 61, 66–74. [Google Scholar] [CrossRef]
  35. Gonyela, O.; Peter, X.; Dewar, J.B.; Van Der Westhuyzen, C.; Steenkamp, P.; Fouché, G. Cycloartanol and Sutherlandioside C peracetate from Sutherlandia frutescens and their immune potentiating effects. Nat. Prod. Res. 2019, 33, 1968–1976. [Google Scholar]
  36. Van Der Walt, N.B.; Zakeri, Z.; Cronjé, M.J. The induction of apoptosis in A375 malignant melanoma cells by Sutherlandia frutescens. Evid. Based Complement. Alternat. Med. 2016, 2016, 4921067. [Google Scholar] [CrossRef]
  37. Van Wyk, B.E.; Gericke, N. Peoples Plants: A Guide to Useful Plants of Southern Africa; Briza Publications: Pretoria, South Africa, 2000. [Google Scholar]
  38. Skerman, N.B.; Joubert, A.M.; Cronjé, M.J. The apoptosis inducing effects of Sutherlandia spp. extracts on an oesophageal cancer cell line. J. Ethnopharmacol. 2011, 137, 1250–1260. [Google Scholar] [CrossRef]
  39. Leisching, G.; Loos, B.; Nell, T.; Engelbrecht, A.-M. Sutherlandia frutescens treatment induces apoptosis and modulates the PI3-kinase pathway in colon cancer cells. S. Afr. J. Bot. 2015, 100, 20–26. [Google Scholar] [CrossRef]
  40. Stander, B.A.; Marais, S.; Steynberg, T.J.; Theron, D.; Joubert, F.; Albrecht, C.; Joubert, A.M. Influence of Sutherlandia frutescens extracts on cell numbers, morphology, and gene expression in MCF-7 cells. J. Ethnopharmacol. 2007, 112, 312–318. [Google Scholar] [CrossRef]
  41. Gouws, C.; Smit, T.; Willers, C.; Svitina, H.; Calitz, C.; Wrzesinski, K. Anticancer potential of Sutherlandia frutescens and Xysmalobium undulatum in LS180 colorectal cancer mini-tumors. Molecules 2021, 26, 605. [Google Scholar] [CrossRef] [PubMed]
  42. Fernandes, A.C.; Cromarty, D.; Albrecht, C.; Van Rensburg, C.E.J. The antioxidant potential of Sutherlandia frutescens. J. Ethnopharmacol. 2004, 95, 1–5. [Google Scholar] [CrossRef]
  43. Jiang, J.; Chuang, D.Y.; Zong, Y.; Patel, J.; Brownstein, K.; Lei, W.; Lu, H.; Simonyi, A.; Gu, Z.; Cui, J.; et al. Sutherlandia frutescens ethanol extracts inhibit oxidative stress and inflammatory responses in neurons and microglial cells. PLoS ONE 2014, 9, e89748. [Google Scholar] [CrossRef] [PubMed]
  44. Tobwala, S.; Fan, W.; Hines, C.J.; Folk, W.R.; Ercal, N. Antioxidant potential of Sutherlandia frutescens and its protective effects against oxidative stress in various cell cultures. BMC Complement. Altern. Med. 2014, 14, 271. [Google Scholar] [CrossRef] [PubMed]
  45. Lei, W.; Browning, J.D.; Eichen, P.A.; Brownstein, K.J.; Folk, W.R.; Sun, G.Y.; Lubahn, D.B.; Rottinghaus, G.E.; Fritsche, K.L. Unveiling the anti-inflammatory activity of Sutherlandia frutescens using murine macrophages. Int. Immunopharmacol. 2015, 29, 254–262. [Google Scholar] [CrossRef]
  46. Grunz-Borgmann, E.; Mossine, V.; Fritsche, K.; Parrish, A.R. Ashwagandha attenuates TNF-α- and LPS-induced NF-κB activation and CCL2 and CCL5 gene expression in NRK-52E cells. BMC Complement. Altern. Med. 2015, 15, 434. [Google Scholar] [CrossRef]
  47. Harnett, S.M.; Oosthuizen, V.; Venter, M.V. Anti-HIV activities of organic and aqueous extracts of Sutherlandia frutescens and Lobostemon trigonus. Phytomedicine 2004, 96, 113–119. [Google Scholar] [CrossRef]
  48. Williams, S.; Roux, S.; Koekemoer, T.; Venter, M.; Van Dealtry, G. Sutherlandia frutescens prevents changes in diabetes-related gene expression in a fructose-induced insulin-resistant cell model. J. Ethnopharmacol. 2013, 146, 482–489. [Google Scholar] [CrossRef]
  49. Enogieru, A.B.; Omoruyi, S.I.; Ekpo, O.E. Aqueous leaf extract of Sutherlandia frutescens attenuates ROS-induced apoptosis and loss of mitochondrial membrane potential in MPP+-treated SH-SY5Y cells. Trop. J. Pharm. Res. 2020, 19, 549–555. [Google Scholar] [CrossRef]
  50. Camille, N.; Dealtry, G. Regulation of M1/M2 macrophage polarization by Sutherlandia frutescens via NFkB and MAPK signaling pathways. S. Afr. J. Bot. 2018, 116, 42–51. [Google Scholar] [CrossRef]
  51. Lei, W.; Browning, J.D.; Eichen, P.A.; Lu, C.H.; Mossine, V.V.; Rottinghaus, G.E.; Folk, W.R.; Sun, G.Y.; Lubahn, D.B.; Fritsche, K.L. Immuno-stimulatory activity of a polysaccharide-enriched fraction of Sutherlandia frutescens occurs by the toll-like receptor-4 signaling pathway. J. Ethnopharmacol. 2015, 172, 247–253. [Google Scholar] [CrossRef] [PubMed]
  52. Müller, A.C.; Patnala, S.; Kis, O.; Bendayan, R.; Kanfer, I. Interactions between phytochemical components of Sutherlandia frutescens and the antiretroviral atazanavir in vitro: Implications for absorption and metabolism. J. Pharm. Pharm. Sci. 2012, 15, 221–233. [Google Scholar] [CrossRef] [PubMed]
  53. Fasinu, P.S.; Gutmann, H.; Schiller, H.; James, A.; Bouic, P.J. The potential of Sutherlandia frutescens for herb-drug interaction. Drug Metab. Dispos. 2012, 41, 488–497. [Google Scholar] [CrossRef] [PubMed]
  54. Stander, A.; Marais, S.; Stivaktas, V.; Vorster, C.; Albrecht, C.; Lottering, M.L.; Joubert, A.M. In vitro effects of Sutherlandia frutescens water extracts on cell numbers, morphology, cell cycle progression, and cell death in tumorigenic and non-tumorigenic epithelial breast cell lines. J. Ethnopharmacol. 2009, 124, 45–60. [Google Scholar] [CrossRef]
  55. Phulukdaree, A.; Moodley, D.; Chuturgoon, A.A. The effects of Sutherlandia frutescens extracts in cultured renal proximal and distal tubule epithelial cells. S. Afr. J. Sci. 2010, 106, 54–58. [Google Scholar] [CrossRef]
  56. Chinkwo, K.A. Sutherlandia frutescens extracts can induce apoptosis in cultured carcinoma cells. J. Ethnopharmacol. 2005, 98, 163–170. [Google Scholar] [CrossRef]
  57. Ngcobo, M.; Gqaleni, N.; Chelule, P.K.; Serumula, M.; Assounga, A. Effects of Sutherlandia frutescens extracts on normal t-lymphocytes in vitro. Afr. J. Tradit. Complement. Altern. Med. 2012, 9, 73–80. [Google Scholar] [CrossRef]
  58. Ntuli, S.S.B.N.; Gelderblom, W.C.A.; Katerere, D.R. The mutagenic and antimutagenic activity of Sutherlandia frutescens extracts and marker compounds. BMC Complement. Altern. Med. 2018, 18, 93. [Google Scholar] [CrossRef]
  59. Prevoo, D.; Swart, P.; Swart, A.C. The influence of Sutherlandia frutescens on adrenal steroidogenic cytochrome P450 enzymes. J. Ethnopharmacol. 2008, 118, 118–126. [Google Scholar] [CrossRef]
  60. Katerere, D.R.; Eloff, J.N. Antibacterial and antioxidant activity of Sutherlandia frutescens (Fabaceae), a reputed anti-HIV/AIDS phytomedicine. Phytother. Res. 2005, 19, 779–781. [Google Scholar] [CrossRef]
  61. Chadwick, W.A.; Roux, S.; van de Venter, M.; Louw, J.; Oelofsen, W. Anti-diabetic effects of Sutherlandia frutescens in Wistar rats fed a diabetogenic diet. J. Ethnopharmacol. 2007, 109, 121–127. [Google Scholar] [CrossRef] [PubMed]
  62. Minocha, M.; Mandava, N.K.; Kwatra, D.; Pal, D.; Folk, W.R.; Earla, R.; Mitra, A.K. Effect of short-term and chronic administration of Sutherlandia frutescens on the pharmacokinetics of nevirapine in rats. Int. J. Pharm. 2011, 413, 44–50. [Google Scholar] [CrossRef] [PubMed]
  63. Omolaoye, T.S.; Skosana, B.T.; Du Plessis, S.S. The effect of Aspalathus linearis, Cyclopia intermedia, and Sutherlandia frutescens on sperm functional parameters of healthy male Wistar rats. Front. Physiol. 2023, 14, 1211227. [Google Scholar] [CrossRef] [PubMed]
  64. Vasaikar, N.; Mahajan, U.; Patil, K.R.; Suchal, K.; Patil, C.R.; Ojha, S.; Goyal, S.N. D-pinitol attenuates cisplatin-induced nephrotoxicity in rats: Impact on pro-inflammatory cytokines. Chem.-Biol. Interact. 2018, 290, 6–11. [Google Scholar] [CrossRef]
  65. Fowler, M.W.; John, E. L-canavanine: A naturally occurring toxic amino acid and its effects on human health. Toxicol. Lett. 2019, 311, 85–92. [Google Scholar]
  66. Steenkamp, V.; Gouws, M.C. Cytotoxicity of six South African medicinal plant extracts used in the treatment of cancer. S. Afr. J. Bot. 2006, 72, 630–633. [Google Scholar] [CrossRef]
  67. BioRender.com. Available online: https://www.biorender.com (accessed on 10 May 2025).
  68. Dwarka, D.; Agoni, C.; Mellem, J.J.; Soliman, M.E.; Baijnath, H. Identification of potential SARS-CoV-2 inhibitors from South African medicinal plant extracts using molecular modeling approaches. S. Afr. J. Bot. 2020, 133, 273–284. [Google Scholar] [CrossRef]
  69. Akindele, A.J.; Sowemimo, A.; Agunbiade, F.O.; Sofidiya, M.O.; Awodele, O.; Ade-Ademilua, O.; Orabueze, I.; Ishola, I.O.; Ayolabi, C.I.; Salu, O.B.; et al. Bioprospecting for Anti-COVID-19 Interventions from African Medicinal Plants: A Review. Nat. Prod. Commun. 2022, 17, 1–24. [Google Scholar] [CrossRef]
  70. Zonyane, S.; Fawole, O.A.; la Grange, C.; Stander, M.A.; Opara, U.L.; Makunga, N.P. The implication of chemotypic variation on the antioxidant and anticancer activities of Sutherlandia frutescens (L.) R.Br. (Fabaceae) from different geographic locations. Antioxidants 2020, 9, 152. [Google Scholar] [CrossRef]
  71. Na, H.-K.; Mossanda, K.S.; Lee, J.-Y.; Surh, Y.-J. Inhibition of phorbol ester-induced COX-2 expression by some edible African plants. Biofactors 2004, 21, 149–153. [Google Scholar] [CrossRef]
  72. Kisten, N. The immune-modulating activity of Sutherlandia frutescens. Master’s Thesis, University of the Western Cape, Cape Town, South Africa, 2020. [Google Scholar]
  73. Faleschini, M.T.; Myer, M.S.; Harding, N.; Fouchè, G. Chemical profiling with cytokine stimulating investigations of Sutherlandia frutescens L. R. (Br.) (Fabaceae). S. Afr. J. Bot. 2013, 85, 48–55. [Google Scholar] [CrossRef]
  74. Choi, M.S.; Lee, W.H.; Kwon, E.Y.; Kang, M.A.; Lee, M.K.; Park, Y.B.; Jeon, S.M. Effects of soy pinitol on the pro-inflammatory cytokines and scavenger receptors in oxidized low-density lipoprotein-treated THP-1 macrophages. J. Med. Food 2007, 10, 594–601. [Google Scholar] [CrossRef] [PubMed]
  75. Sivakumar, S.; Palsamy, P.; Subramanian, S.P. Impact of D-pinitol on the attenuation of proinflammatory cytokines, hyperglycemia-mediated oxidative stress, and protection of kidney tissue ultrastructure in streptozotocin-induced diabetic rats. Chem. Biol. Interact. 2010, 188, 237–245. [Google Scholar] [CrossRef] [PubMed]
  76. Leu, G.; Lin, T.; John, T.A. Anti-HCV activities of selected polysaturated fatty acids. Biochem. Biophys. Res. Commun. 2004, 318, 275–280. [Google Scholar] [CrossRef]
  77. McGaw, L.J.; Jäger, A.J.; Van Staden, J. Antibacterial effects of fatty acids and related compounds from plants. S. Afr. J. Bot. 2002, 68, 417–423. [Google Scholar] [CrossRef]
  78. Obonyo, M.; Zhang, L.; Thamphiwatana, S.; Pornpattananangkul, D.; Fu, V.; Zhang, L. Antibacterial activities of liposomal linolenic acids against antibiotic-resistant Helicobacter pylori. Mol. Pharm. 2012, 9, 2677–2685. [Google Scholar] [CrossRef]
  79. Korb, V.C.; Moodley, D.; Chuturgoon, A.A. Apoptosis-promoting effects of Sutherlandia frutescens extracts on normal human lymphocytes in vitro. S. Afr. J. Sci. 2010, 106, 64–69. [Google Scholar] [CrossRef]
  80. Engelbrecht, A.; Smith, C.; Neethling, I.; Thomas, M.; Mattheyse, M.; Myburgh, K.H.; Ellis, B. Daily brief restraint stress alters signaling pathways and induces atrophy and apoptosis in rat skeletal muscle. Stress 2010, 13, 132–141. [Google Scholar] [CrossRef]
  81. Mombereau, C.; Kaupmann, K.; Froestl, W.; Sansig, G.; Van Der Putten, H.; Cryan, J.F. Genetic and pharmacological evidence of a role for GABAB receptors in the modulation of anxiety- and antidepressant-like behavior. Neuropsychopharmacology 2004, 29, 1050–1062. [Google Scholar] [CrossRef]
  82. Green, M.H. Method of Treating Viral Infections with Amino Acid Analogs. U.S. Patent No. 5,110,600, 25 January 1988. [Google Scholar]
  83. Africa, L.; Smith, C. Sutherlandia frutescens may exacerbate HIV-associated neuroinflammation. J. Negat. Results Biomed. 2015, 14, 14. [Google Scholar] [CrossRef]
  84. Zonyane, S.; Chen, L.; Xu, M.J.; Gong, Z.N.; Xu, S.; Makunga, N.P. Geographic-based metabolomic variation and toxicity analysis of Sutherlandia frutescens L. R.Br.—An emerging medicinal crop in South Africa. Ind. Crops Prod. 2019, 133, 414–423. [Google Scholar] [CrossRef]
  85. Mackenzie, J.; Koekemoer, T.; Venter, M.; Van de Dealtry, G. Sutherlandia frutescens limits the development of insulin resistance by decreasing plasma free fatty acid levels. Phytother. Res. 2009, 23, 1609–1614. [Google Scholar] [CrossRef] [PubMed]
  86. Bates, S.H.; Jones, R.B.; Bailey, C.J. Insulin-like effect of pinitol. Br. J. Pharmacol. 2000, 130, 1944–1948. [Google Scholar] [CrossRef] [PubMed]
  87. Grandi, M.; Roselli, L.; Vernay, M. Lessertia (Sutherlandia frutescens) and fatigue during cancer treatment. Phytothérapie 2005, 3, 110–113. [Google Scholar] [CrossRef]
  88. Chaffy, N.; Stokes, T. Aids herbal therapy. Trends Plant Sci. 2002, 7, 57. [Google Scholar]
  89. Wilson, D.; Goggin, K.; Williams, K.; Gerkovich, M.M.; Gqaleni, N.; Syce, J.; Bartman, P.; Johnson, Q.; Folk, W.R.; Doherty, T.M. Consumption of Sutherlandia frutescens by HIV-seropositive South African adults: An adaptive double-blind randomized placebo-controlled trial. PLoS ONE 2015, 10, e0128522. [Google Scholar] [CrossRef]
  90. Fang, X.J.; Jiang, H.; Zhu, Y.Q.; Zhang, L.Y.; Fan, Q.H.; Tian, Y. Doxorubicin induces drug resistance and expression of the novel CD44st via NF-κB in human breast cancer MCF-7 cells. Oncol. Rep. 2014, 31, 2735–2744. [Google Scholar] [CrossRef]
  91. Liebmann, J.E.; Cook, J.A.; Lipschultz, C.; Teague, D.; Fisher, J.; Mitchell, J.B. Cytotoxic studies of paclitaxel (Taxol®) in human tumor cell lines. Br. J. Cancer 1993, 68, 1104–1109. [Google Scholar] [CrossRef]
  92. Sieniawska, E. Activities of tannins from in vitro studies to clinical trials. Nat. Prod. Commun. 2015, 10, 1877–1884. [Google Scholar] [CrossRef]
  93. De Clercq, E. Antiviral drugs in current clinical use. J. Clin. Virol. 2004, 30, 115–133. [Google Scholar] [CrossRef]
  94. Paintsil, E.; Cheng, Y.C. Antiviral agents. In Encyclopedia of Microbiology, 3rd ed.; Schaechter, M., Ed.; Academic Press: San Diego, CA, USA, 2009; pp. 223–257. [Google Scholar]
  95. Wassner, C.; Bradley, N.; Lee, Y. A review and clinical understanding of tenofovir: Tenofovir disoproxil fumarate versus tenofovir alafenamide. J. Int. Assoc. Provid. AIDS Care 2020, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
  96. Okoye, A.A.; Picker, L.J. CD4+ T-cell depletion in HIV infection: Mechanisms of immunological failure. Immunol. Rev. 2013, 254, 54–64. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, Y.; Lui, M.; Lu, Q.; Farrell, M.; Lappin, J.; Shi, J.; Lu, L.; Bao, Y. Global prevalence and burden of HIV-associated neurocognitive disorder: A meta-analysis. Neurology 2020, 95, e2610–e2621. [Google Scholar] [CrossRef] [PubMed]
  98. Persistance Market Research. Sutherlandia Extract Market. Available online: https://www.persistencemarketresearch.com/market-research/sutherlandia-extract-market.asp (accessed on 7 April 2024).
  99. Marais, J.F.; Van Jaarsveld, J.J.; Laporta, J.C.H. Sutherlandia Extract and the Use Thereof in the Manufacture of a Medicament. Patent WO 2022/130222 A1, 23 June 2022. [Google Scholar]
  100. Visser, J.S. A Pharmaceutical. Composition. Patent WO2012/063225 A1, 18 May 2012. [Google Scholar]
Figure 1. Lessertia frutescens [23].
Figure 1. Lessertia frutescens [23].
Plants 14 02086 g001
Figure 2. Traditional uses of L. frutescens range from relaxation to healing.
Figure 2. Traditional uses of L. frutescens range from relaxation to healing.
Plants 14 02086 g002
Figure 3. Chemical structures of L. frutescens isolated compounds (113). Created using ChemOffice Pro 2015 (version 15.00).
Figure 3. Chemical structures of L. frutescens isolated compounds (113). Created using ChemOffice Pro 2015 (version 15.00).
Plants 14 02086 g003
Figure 4. Chemical constituents of L. frutescens (1431). Created using ChemOffice Pro 2015 (version 15.00).
Figure 4. Chemical constituents of L. frutescens (1431). Created using ChemOffice Pro 2015 (version 15.00).
Plants 14 02086 g004
Figure 5. Molecular pathways modulated by L. frutescens extracts: Hedgehog, mitochondrial, and PI3K/AKT signalling in cancer models. Created using BioRender.com [67].
Figure 5. Molecular pathways modulated by L. frutescens extracts: Hedgehog, mitochondrial, and PI3K/AKT signalling in cancer models. Created using BioRender.com [67].
Plants 14 02086 g005
Figure 6. Cytotoxic effects of L. frutescens on renal epithelial cells: oxidative stress and mitochondrial dysfunction. Created using BioRender.com [67].
Figure 6. Cytotoxic effects of L. frutescens on renal epithelial cells: oxidative stress and mitochondrial dysfunction. Created using BioRender.com [67].
Plants 14 02086 g006
Figure 7. Inhibitory effects of L. frutescens extracts on inflammatory signalling pathways. Created using BioRender.com [67].
Figure 7. Inhibitory effects of L. frutescens extracts on inflammatory signalling pathways. Created using BioRender.com [67].
Plants 14 02086 g007
Figure 8. Mechanism of action of L. frutescens on HIV-1 reverse transcriptase. Created using BioRender.com [67].
Figure 8. Mechanism of action of L. frutescens on HIV-1 reverse transcriptase. Created using BioRender.com [67].
Plants 14 02086 g008
Figure 9. Inhibition of P450 enzyme activity. Created using BioRender.com [67].
Figure 9. Inhibition of P450 enzyme activity. Created using BioRender.com [67].
Plants 14 02086 g009
Figure 10. Impact of L. frutescens extracts on HIV-induced neuroinflammation. Created using BioRender.com [67].
Figure 10. Impact of L. frutescens extracts on HIV-induced neuroinflammation. Created using BioRender.com [67].
Plants 14 02086 g010
Figure 11. L. frutescens clinical trials outcomes. Created using BioRender.com [67].
Figure 11. L. frutescens clinical trials outcomes. Created using BioRender.com [67].
Plants 14 02086 g011
Figure 12. The different forms of L. frutescens in the market (Pictures sourced from the South African local market).
Figure 12. The different forms of L. frutescens in the market (Pictures sourced from the South African local market).
Plants 14 02086 g012
Table 1. The IUPAC names of the sutherlandiosides E-H isolated by Fu [33] and Tchegnitegni et al. [34].
Table 1. The IUPAC names of the sutherlandiosides E-H isolated by Fu [33] and Tchegnitegni et al. [34].
Compounds Fu [33]Tchegnitegni et al. [34]
Sutherlandioside E24,25-O-β-D-diglucopyranosyl-3S, 24S,25-trihydroxy, 2(1-10)-abeo-9, 10R-seco-cycloartanoic acid (20)7S,24S,25-trihydroxy-9, 10R-seco-9,19-cyclolanost-2(3), 9(11)-diene-25-O-β-D-glucopyranoside (22)
Sutherlandioside F24,25-O-β-D-diglucopyranosyl 3R, 24S,25-trihydroxy, 2(1-10)-abeo-9, 10R-seco-cycloartanoic acid (21)1S,3R,7S,24S,25-pentahydroxycycloartan-11-one-25-O-β-D-glucopyranoside (23)
Sutherlandioside G(3R,7S,24S,25)-[3-O-β-D-glucopyranosyl]-tetrahydroxycycloartan-1-one-25-O-β-D-glucopyranoside (18)1S,3R,7S,24S,25-pentahydroxycycloartan-11-one-24-O-β-D-glucopyranosyl-25-O-β-D-glucopyranoside (24)
Sutherlandiosde H3R,24S,25-trihydroxycycloartane-1,11-
dione-24, 25-O-β-D-diglucopyranoside (19)
3R,24S,25-trihydroxycycloartan-1-one-25-O-β-D-glucopyranoside (25)
Table 2. Ethnopharmacological and pharmacological properties of L. frutescens extracts, fractions, and compounds in in vitro studies (whole plant extracts considered when specific plant parts are not mentioned).
Table 2. Ethnopharmacological and pharmacological properties of L. frutescens extracts, fractions, and compounds in in vitro studies (whole plant extracts considered when specific plant parts are not mentioned).
Pharmacological ActivityExtract/CompoundExperimental ModelsTested ConcentrationsExperimental OutcomesReferences
Anticancer70% ethanol (tablet)MDA-MB-468, HL-60, MCF-7, and Jurkat cellsIC50: 0.91–0.55 mg/mLInhibited growth of MCF7 (0.55 mg/mL), MDA-MB-468 (0.68 mg/mL), Jurkat (0.91 mg/mL), and HL60 (0.68 mg/mL)[9]
A-375, Colo-800 cells cells0.625 mg/mLInhibited proliferation [36]
HDFα cells0.3 mg/mLReduced HDFα viability to 19% after 72 h
70% ethanolPC3, LNCaP, and TRAMP-C2 cells100–200 µg/mL (IC50)Suppressed the growth of PC3 (167), LNCaP (200), and TRAMP-C2 (100) while inhibiting Gli/Hh signalling activity by downregulating Gli1 and PTCH1 gene expression in TRAMP-C2 and PC3 cells[10]
SNO cells2.5 and 5 mg/mLReduced ATP levels; apoptotic effect; decreased caspase 3/7 levels[38]
CaCo-2 cells2.73 mg/mLInhibited the PI3-K/Akt pathway by decreasing the phosphorylation of p85, p110, Akt (Ser473 and Thr308), and PTEN
Promoted apoptosis by increasing PARP cleavage and cleaved caspase-9 levels while significantly reducing total Bax and c-IAP levels
[39]
70% ethanolic (leaves and twigs) MCF-7 cells1.5 mg/mLInhibited the proliferation of MCF-7 cells after 72 h of exposure[40]
Sutherlandioside B and DShh Light II cells10 µg/mLInhibited Gli-reporter activity by 22% and 89%, respectively[10]
AqueousLS180 cells2.63 mg/mL (IC50)Reduced cell growth, viability, ATP, and AK levels relative to protein content[41]
CYP3A4 and CYP2D6 enzymes2.63 mg/mL (IC50) Inhibited the gene expression of CYP3A4 and CYP2D6 enzymes
AntioxidantAqueousFMLP-stimulated neutrophils10 µg/mLDecreased luminol and lucigenin-enhanced chemiluminescence response[42]
Oxidant-scavenging in cell-free systems10 and 0.62 µg/mLInhibited superoxide-induced chemiluminescence at 10 µg/mL and horseradish peroxidase/hydrogen peroxide-induced chemiluminescence at 0.62 µg/mL
L-canavanine, D-PinitolRAW 264.7 cells0.5 mM (L-c), 10 mM (D-p)Inhibited LPS-induced NO secretion without reducing cell numbers[9]
70% ethanolPrimary rat cortical neurons0.1–7.5 µg/mLInhibited NMDA-induced ROS production without altering viability[43]
AqueousCHO, HepaRG, and A549500 µg/mLProtected cells from t-BHP-induced oxidative stress by scavenging ROS, preserving GSH/GSSG levels[44]
1 mg/mLPotent scavenger of hydroxyl, superoxide, and hydrogen peroxide radicals
Anti-inflammatory70% ethanolBV-2 and HAPI microglial cells0.1–80 µg/mLInhibited IFN-γ-induced p-ERK1/2 and p-STAT-1α expression as well as NO and filopodia production (BV-2)
Inhibited LPS+ IFN-γ induced ROS and NO production as well as iNOS expression (BV-2 and HAPI)
[43]
EthanolicRAW 264.7 cells200 µg/mLReduced NO, iNOS, IL-6, and TNF-α production; inhibited ERK1/2, STAT1-α, and NF-κB activation[45]
Sutherlandioside B-enrichedRAW 264.7 cells200 µg/mLReduced ROS induced by LPS and IFN-γ[45]
Aqueous (leaves)NRK-52E cells0.4 mg/mLPartially reduced TNF-α induced chemokine CCL5 expression[46]
AntidiabeticDichloromethane (leaves)α- and β-glucosidase enzymes0.2 mg/mLSignificantly inhibited α- and β-glucosidase enzymes[47]
AqueousHepatocytes12.5 µg/mLPrevented insulin resistance[48]
NeuroprotectionAqueousMPP+-induced toxicity (SH-SY5Y cells)20 µg/mLReduced ROS production[49]
Immune ModulatoryAqueous and ethanolic (leaves)RAW 264.7 cells200 µg/mLInhibited LPS-induced ERK1/2 and p38 phosphorylation; reduced NO, ROS, TNF-α, IL-6, GM-CSF, and G-CSF[50]
100 µg/mLReduced LPS-stimulated NO and ROS production
50 µg/mLReduced LPS-induced production of IL-1α
100, 150 and 200 µg/mLInhibited NF-κB activation by attenuating NF-κB p65 subunit phosphorylation on the Ser 536 residue
Ethanol150 and 100 µg/mLReduced CD86 expression and inhibited COX-2
100 µg/mLincreased the CD206 cell surface marker expression
Polysaccharide-enriched fraction 200 µg/mLActivated macrophages via TLR4 receptors and NF-κB signalling pathway[51]
Anti-HIVAqueous (leaves)HIV-1 RT enzyme0.2 mg/mLInhibited HIV-1 reverse transcriptase enzyme by ≥50%[47]
Methanolic and aqueousHuman liver microsomes10 mg/mLInhibited the metabolism of atazanavir[52]
AqueousCaCo-2 cells10 mg/mLDecreased atazanavir accumulation, bioavailability, and absorption
The triterpenoid glycoside-enriched fractionCaCo-2 cells500 µg/mLIncreased atazanavir accumulation and enhanced its bioavailability and absorption
Human liver microsomes Decreased the atazanavir present (p < 0.001) in human liver microsomes
40% aqueous methanolP450 enzymesIC50 ranging from 17–160 μg/mLInhibited CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP3A4/5, and CYP3A4/5 with IC50 values of 41, 160, 20, 22.4, 23, 35.9, 17.5, and 28.3 μg/mL, respectively[53]
Hepatocytes100 µg/mLReduced midazolam clearance by 40% by delaying the production of midazolam metabolites
LLC-PK1 cells stably transfected with human P-gp324.8 µg/mL (IC50)Inhibit P-gp
Human embryonic kidney 293 cells stably transfected with human OATP1B1 or OATP1B3IC50Inhibit OATP1B1 (10.4 µg/mL) and OATP1B3 (6.6 µg/mL)
Cytotoxic70% ethanolMCF-7 and MCF-12A10 mg/mLReduced cell growth and induced apoptosis in MCF-7 and MCF-12A[54]
AqueousMDBK and LLC-PK1 cells6 mg/mLDisrupted mitochondrial integrity, promoted apoptosis[55]
CHO and cervical neoplastic cells3.5 mg/mLActivated apoptosis[56]
70% ethanol and aqueous (tablet)Normal T-lymphocytes2.5 mg/mLInduced necrosis, depleted ATP, inhibited caspase 3/7 activity, and caused DNA fragmentation[57]
AntimutagenicEthyl acetateTA97a, TA98, TA100, and TA102 strains5, 10, 20% (w/w)Exhibited antimutagenic effect against multiple strains[58]
L-arginine, GABA, D-PinitolTA97a, TA98, TA100, and TA102 strains0.05–0.49 MExhibited antimutagenic activity against all four strains
Pro-mutagenicMethanolTA98 and TA100 strains10, 25, 50% (w/w)Showed pro-mutagenic potential in TA98 with 2-acetamidofluorene and TA100 with aflatoxin B1
Anti-tuberculosisDichloromethane–methanol (1:1)Mycobacterium tuberculosisIC50: 0.1–5.1 μg/mLInhibited the shikimate kinase enzyme
[11]
α-Linolenic acidMycobacterium tuberculosis3.7 µg/mLInhibited the shikimate kinase enzyme
Different DCM: MeOH fractionsMycobacterium tuberculosis0.3–94.3 µg/mLInhibited the shikimate kinase enzyme at varying IC50 values
Anti-stressMethanol, chloroform, and aqueousOvine adrenal mitochondria and microsomes (0.78 mM P450 in binding studies and 0.33 mM P450 in conversion)2.4 (50 µL of 48 mg/mL) and 4.1 mg (50 µL of 82 mg/mL)Decreased the binding of DOC, PROG, and PREG, as well as the conversion of PROG and PREG (methanol and chloroform, 4.1 mg)
Inhibited substrate binding to CYP21 and CYP11B1 (aqueous, 2.4 mg)
[26]
Triterpenoid fractionP450 enzymes CYP17 and CYP21 enzymes0.6 and 1.5 mg/mLInhibited the binding of PROG (0.6 mg/mL) and PREG (1.5 mg/mL)[59]
Methanol and aqueous2.4 and 4.1 mg/mL
Methanol (4.1 mg/mL) and aqueous (2.4 mg/mL) extracts inhibited the binding of PROG
AqueousAdrenocortical microsomes2.4 mg/mLInhibited PROG metabolism and the formation of DOC, 17-OH-PROG and deoxycortisol (46%)
COS-1 cellsInhibited PREG and PROG metabolism and the formation of the hydroxysteroid intermediates than DHEA and A4
MethanolCOS-1 cells2.6 mg/mLInhibited PROG binding to CYP17A1 and CYP21A2[12]
Human H295R adrenal cells1 mg/mLDecreased total steroid production under basal steroidogenesis and forskolin-stimulated steroidogenesis conditions
Inhibited CYP17A1 and CYP11B1 and significantly reduced PROG, DOC, CORT, 17OH-PREG, 16OH-PROG, 11-DHC, 11OHA4, and A4 levels, while increasing DHEAS
COS-1 cells0.5–0.75 mg/mLAntagonised the effects of ALDO via the MR
Significantly repressed the IL-6 promoter after stimulation with PMA (10 ng/mL)
Sutherlandioside BCOS-1 cells0.5 and 0.75 mg/mLSuppressed NF-κB-driven gene expression while antagonising the effects of ALDO via the MR[12]
COS-1 cells10 and 30 µMInhibited CYP17A1 activity toward PREG and PROG, as well as 3β-HSD2 activity toward PROG, and acted as selective glucocorticoid receptor agonists
In H295R cells30 µMDecreased CORT, A4, 11OH-A4, and 16OH-PROG, while increasing 11-DHC
AntibacterialHexaneEnterococcus faecali, Escherichia coli, and Staphylococcus aureusIC50 varying from 0.31 to 2.5 mg/mLInhibited bacteria with IC50 of 2.50 (Ef), 1.25 (Ec), and 0.31(Sa) mg/mL[60]
Table 3. Ethnopharmacological and pharmacological properties of L. frutescens extracts and compounds in in vivo studies.
Table 3. Ethnopharmacological and pharmacological properties of L. frutescens extracts and compounds in in vivo studies.
Pharmacological Activity Extract/Compound Experimental Models Tested Concentrations Experimental Outcomes References
Anti-analgesicAqueous (shoot)Hot-plate and acetic acid test models of pain in mice50–800 mg/KgProduced analgesic effects against thermally and chemically induced nociceptive pain stimuli in mice[3]
Anti-inflammatoryFresh egg albumin-induced pedal oedema50–800 mg/KgInhibited fresh egg albumin-induced acute inflammation[3]
AntidiabeticAqueous (shoot)Streptozotocin-induced hyperglycemia in mice50–800 mg/KgInhibited streptozotocin-induced hyperglycemia[3]
AqueousWistar rats fed a diabetogenic diet0.01 mL/g rat weightIncreased glucose uptake into muscle and adipose tissue while significantly decreasing intestinal glucose uptake[61]
Anti-convulsionAqueous (shoot)Streptozotocin (PTZ)-induced seizures in mice50–800 mg/kgProtected the mice against PTZ-induced seizures[27]
Picrotoxin (PCT)-induced seizures in mice50–800 mg/kgProtected the mice against PCT-induced seizures[27]
Anti-HIV70% EthanolRat hepatic and intestinal tissues12 mg/kgIncreased intestinal and hepatic rat CYP3A2 expression levels after 5 days of exposure[62]
Rats6 mg/kgAltered the pharmacokinetics of nevirapine after 5 days of chronic exposure by reducing the AUC0–inf and Cmax
Antioxidant/SpermatotoxicMethanolWistar rats2 mg/mLRapid progressive motility decreased, while slow-moving spermatozoa, catalase activity, and SOD activity significantly increased. The rise in SOD activity was associated with a reduction in MDA levels[63]
Anti-nephrotoxicityD-pinitolCisplatin-induced nephrotoxicity in mice10–40 mg/kg/dayPrevented alterations in renal biomarkers, urine creatinine, serum blood urea nitrogen, and NO levels; improved histopathological alterations by preventing severe necrosis[64]
Cisplatin-induced nephrotoxicity in mice10, 20, and 40 mg/kg/dayAltered cisplatin-induced changes in inflammatory markers by decreasing the levels of TNF-α, IL-1β, IL-6, and NO
Table 4. Pharmacological properties of L. frutescens extracts in clinical trial studies.
Table 4. Pharmacological properties of L. frutescens extracts in clinical trial studies.
Pharmacological ActivityExtractsExperimental ModelTested ConcentrationsExperimental OutcomesReferences
AnticancerAqueous16 cancer patients (11 men and 5 women)600 mg/dayDecreased fatigue in cancer patients (oral consumption).[87]
Non-cytotoxicleaf powder (capsules)25 healthy adults800 mg/dayImproved appetite in the treatment group. Overall, healthy adults tolerated 800 mg/d of Sutherlandia leaf powder well for three months.[5]
Anti-HIVLeaf powder107 participants2400 mg/day (1200 mg twice daily)did not alter the viral load, and the CD4 T-lymphocyte count remained the same in the two arms.[89]
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

Ndjoubi, K.O.; Sharma, R.; Hussein, A.A. Phytochemistry, Ethnopharmacology, and Pharmacology of Lessertia frutescens (Cancer Bush): A Comprehensive Review. Plants 2025, 14, 2086. https://doi.org/10.3390/plants14142086

AMA Style

Ndjoubi KO, Sharma R, Hussein AA. Phytochemistry, Ethnopharmacology, and Pharmacology of Lessertia frutescens (Cancer Bush): A Comprehensive Review. Plants. 2025; 14(14):2086. https://doi.org/10.3390/plants14142086

Chicago/Turabian Style

Ndjoubi, Kadidiatou O., Rajan Sharma, and Ahmed A. Hussein. 2025. "Phytochemistry, Ethnopharmacology, and Pharmacology of Lessertia frutescens (Cancer Bush): A Comprehensive Review" Plants 14, no. 14: 2086. https://doi.org/10.3390/plants14142086

APA Style

Ndjoubi, K. O., Sharma, R., & Hussein, A. A. (2025). Phytochemistry, Ethnopharmacology, and Pharmacology of Lessertia frutescens (Cancer Bush): A Comprehensive Review. Plants, 14(14), 2086. https://doi.org/10.3390/plants14142086

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