Keywords
anticancer - cytotoxic - Fabaceae - herbal medicine - marketing -
Sutherlandia frutescens
- traditional remedy
Introduction
Sutherlandia (syn. Lessertia) frutescens (L.) R.Br. ex W. T.Aiton (Fabaceae), colloquially known as the ‘cancer bush’ or ‘Kankerbos’
(Afrikaans), is a popular herbal remedy in South Africa, revered for its medicinal
properties and, as the common name implies, for its purported anticarcinogenic properties.
This drought-resistant shrub belongs to the Fabaceae family, the third-largest family
of flowering plants, and is widely distributed throughout the drier regions in the
Western, Eastern, and Northern Cape and parts of the KwaZulu-Natal provinces of South
Africa, as well as other parts of southern Africa, including Botswana, Zimbabwe, and
Namibia [1]. Despite the widespread use and commercial availability of S. frutescens, concrete scientific data surrounding its mechanism of action against the proliferation
of carcinomic cells remains limited, and the evidence for its efficacy as a prophylaxis
and/or treatment for cancer
is seemingly inconclusive [2]. Due to these uncertainties, it was decided to collate all available scientific
data regarding the antiproliferative effects of the genera Sutherlandia and Lessertia and critically evaluate the presented data to ascertain if there is any scientific
merit to the anticancer health claims.
An updated overview of this very popular herb is provided here. This review is divided
into three sections: the first addresses general topics such as botany and traditional
use, and the second briefly covers the literature on its phytochemistry and toxicity,
whilst the bulk of this review covers its antiproliferative properties and a critical
appraisal of published data. The objective of this review was therefore to collate
all scientific data on the genera Lessertia and Sutherlandia with the main focus, as the vernacular name ‘cancer bush’ implies, being their anticarcinogenic,
antiproliferative, antineoplastic, and cytotoxic activity and to critically evaluate
the data against established guidelines. Recommendations regarding the use of this
popular herb, as well as to identify a direction of future research, are also presented.
Search Strategy
Scientific literature published in English up to December 2024 was retrieved from
scientific databases (Science Direct, Web of Science, Scopus, PubMed, and Google Scholar),
as well as from several (historical) books regarding medicinal plants and ethnobotany.
As an indication of the overall amount of all scientific research that has been conducted
on these two genera, the Web of Science lists only 55 publications using the keyword
‘Lessertia’ and 201 publications using the keyword ‘Sutherlandia’, with the vast majority not applicable to this review.
Botanical Aspects
Sutherlandia frutescens is native, but not limited to, the Cape Floristic Kingdom and is specifically found
in the Fynbos biome, which hosts one of the greatest varieties of plant species in
the world. Classified in the class Magnoliopsida, order Fabales, and genus Sutherlandia, this plant has been suggested for reclassification under the genus Lessertia, based on the suggestion that it has adapted to bird pollination [1]. However, the name Sutherlandia is commonly used on commercial products and colloquially and hence will be used to
describe this genus throughout this manuscript. According to the International Plant
Name Index, the following Sutherlandia species are recognised: S. darumbium Bertero; S. floribunda Carrière; S. foliolata Lange; S. frutescens; S. humilis E.Phillips & R. A.Dyer; S. incana E.Mey.; S. littoralis J. F.Gmel.; S.
microphylla Burch. ex DC.; S. montana E.Phillips & R. A.Dyer; S. speciosa E.Phillips & R. A.Dyer; S. tomentosa Spreng.; and S. vesicaria Spreng.
Sutherlandia frutescens is a medium-sized shrub (roughly 0.2 – 1.5 m in height) with fine silver-green leaves
and red butterfly-shaped flowers, while its seedpods are large, balloon-like, and
slightly reddish, hence the common name ‘balloon pea’ ([Fig. 1]) [1], [3]. Although mostly harvested from the wild, it is also cultivated in community gardens
and commercially. The plant is known by over 25 different common names, many reflecting
its characteristics, such as ‘kalkoenbos’ (turkey bush) and others referring to its
medicinal uses, like ‘Kankerbos’ (cancer bush), ‘insiswa’ (dispels darkness), and
‘lerumo lamadi’ (spear of the blood) [1], [3]. The taxonomy is debated, with various authors classifying the genus differently.
The taxonomy of this genus was reviewed, and for the current review and the sake of
thoroughness, both genera
Lessertia and Sutherlandia and their purported effect on cancer will be detailed [4].
Fig. 1 a Sutherlandia frutescens inflorescences b leaves and flowers c seed pods and d Sutherlandia frutescens shrub (Photos: with permission of Random Harvest Nursery).
The Ethnopharmacology of Lessertia and Sutherlandia
The Ethnopharmacology of Lessertia and Sutherlandia
The first records of its use date back to the Dutch colonists in the Cape, who likely
learned of it through interactions with the indigenous Khoi-San people. The Khoi-San
and Nama traditionally utilised decoctions of Sutherlandia to cleanse wounds and reduce fevers [1]. One of the earliest mentions of S. frutescens and its use as a possible cancer treatment was documented in 1895, when it was noted
that it was commonly used for the treatment of cancer, but a physician concluded that
it was essentially ineffective [5]. A valuable source of information regarding the traditional use of plants in southern
Africa was published in 1932 by Watt and Breyer-Brandwijk, with a second edition published
in 1962 [6]. This resource is not electronically available, and there is a significant risk
that this valuable and insightful source may eventually be lost. Therefore, information
from this
resource should be collated and made available electronically via scientific publications.
Here we present all that is documented regarding the genus Lessertia and Sutherlandia as recorded by [6].
Lessertia annularis Burch. was mentioned as the possible cause of cotyledonosis (‘krimpsiekte’) but this
was later attributed to Cotyledon ventricose Burm. However, L. annularis was stated to be toxic. L. brachystachya DC. was shown to be toxic to cattle during feeding experiments, whereas L. physodes E.&Z. did not show any toxicity. L. argentea Harv. was utilised as an eye lotion but also to alleviate flatulence and colic, and
L. tomentosa DC. was documented to be used by the early settlers as a remedy for ophthalmic conditions.
An infusion of S. frutescens R.Br. var. incana E.Mey. was used orally as a cough remedy and for cancer and externally as a female
hygiene product. It is interesting to note that the infusion induced vomiting when
it was prepared too ‘strong’, but no details regarding the preparation method were
provided. Similarly, a decoction of S. humilis Phill. & Dyer and an infusion of
the bark and leaves were used as a treatment for cancer as well as influenza.
Sutherlandia microphyla Burch. was stated to be very popular and the most commonly used Sutherlandia species throughout South Africa. It was specified that a ‘weak’ infusion or decoction
of the leaves was taken in unlimited amounts to treat a wide variety of ailments,
including stomach complaints, internal cancers, uterine complaints, influenza, liver
disease, rheumatism, inflammations, haemorrhoids, dropsy, and backache. An infusion
was also consumed for amenorrhea and as a cancer prophylactic. The powdered leaf mixed
with syrup was claimed to be soothing for a troublesome cough and was also used as
a tonic to improve appetite and digestion. However, when the infusion was prepared
too ‘strong’, without defining the preparation method, it caused sweating and acted
as a purgative.
The Southern Sotho used the plant as an infusion for the treatment of fever as well
as ‘dropsy of the heart’, whereas the Nama used a decoction to treat wounds, fevers,
consumption (TB), chicken-pox, and a variety of other conditions. In North Africa,
the leaf was used as a tonic and for indigestion and dysentery. A clinical trial was
conducted using an infusion of the bark and the whole plant for the treatment of cancer,
but the results of the trials were negative. The original document published in 1918,
however, only stated that “Cancer bush (Sutherlandia frutescens). A half shrub, two to four feet in height, with scarlet flowers. It is supposed
to cure cancer. Clinical experiments have proved this belief to be entirely without
foundation” [6], [7].
It is claimed that the consumption of half a cup of Sutherlandia infusion or decoction three times daily would aid in the treatment of cancer and
that the intake of a Sutherlandia decoction would prevent cancer metastasis [8], [9]. Improved quality of life of patients suffering from pancreatic, breast, and other
cancer was reported with the administration of a Sutherlandia tonic [10], whereas it was asserted that a correlation exists between the use of Sutherlandia and a reduction in cancer-induced muscle wasting [11]. The traditional use of S. frutescens is categorised under wound healing, anticancer, antiviral, antibacterial, and anti-inflammatory
treatments, along with the alleviation of gastrointestinal-related ailments and mood
disorders (such as anxiety, depression, and mood imbalances). Generally, Sutherlandia is
used as a bitter tonic, tranquiliser, and blood purifier [3].
In South Africa, S. frutescens is widely used by various healers, including herbalists (inyanga), diviners (isangoma),
bush doctors (bossiedokters), Rastafarians, and both alternative and allopathic medicine
practitioners, as well as by the public [1]. While Sutherlandia has long been appreciated as a decorative garden plant for flower arrangements, it
is most renowned for its medicinal properties, particularly as an herbal medicine
to improve the quality of life in HIV/AIDS patients [1], [12]. Several commercial farms currently produce S. frutescens to satisfy the growing demand for this popular herb, which is mainly being sold as
an over-the-counter complementary medicine [1].
Phytochemistry
No comprehensive phytochemical analysis on this species has yet been conducted. The
identified phytochemicals have been reviewed and include free amino acids, non-protein
amino acids (L-canavanine, GABA), the cyclitol, pinitol, flavonoids, flavonoid glycosides,
and cycloartane glycosides [4]. The flavonoid glycosides were identified as sutherlandins A to D [13] and the cycloartane glycosides as sutherlandiosides A – D, with sutherlandioside
B commonly known as SU1 [14], [15]. The sutherlandins and sutherlandiosides are currently considered chemical markers
for the species, and various publications could be found using these phytochemicals
as quality-control marker compounds [3], [16], [17], [18]. Recently, a somewhat more in-depth analysis regarding
the presence of other phytochemicals was conducted, and seven new cycloartane glycosides
(sutherlandiosides E – K) and one oleanan-type saponin were identified [19]. Little information exists as to the antiproliferative activity of these compounds,
and this aspect will be discussed in section 4.2.
Pharmacological Aspects
Toxicity
No serious adverse events have been reported with the use of S. frutescens, and only mild side effects such as dry mouth, diarrhoea, mild diuresis, and dizziness
were described [11]. In 1908, it was stated that typical (effective) dosages for traditional infusions
ranged from 2.5 g to 5.0 g of dried plant material daily [20]. High doses, such as a decoction prepared from 5 g taken daily for over 6 years,
did not result in any reported side effects [4]. Toxicity studies on Sutherlandia have indicated a median lethal dose (LD50) of 1280 ± 71 mg/kg in mice, suggesting the plant is relatively safe in mammals [21]. For commercial preparations, 600 mg of dried leaves per day is generally recommended,
with precautions during pregnancy or lactation. In a safety study in vervet monkeys,
doses up to nine times the recommended amount showed no
significant toxic effects [22]. A clinical trial in 25 healthy adults found that 800 mg of leaf powder daily was
well tolerated, with no serious side effects reported [23]. Some minor physiological changes were observed but remained within normal ranges.
However, caution is advised, as Sutherlandia may interact with antiretroviral medications, diabetes drugs, and tuberculosis therapies
[3], [11], [24].
Antiproliferative Studies
[Tables 1] and [2] provide, to the best of our knowledge, the details of all published antiproliferative
studies conducted using S. frutescens. The source of plant material, plant parts used, and sample preparation methods are
presented in [Table 1], whereas [Table 2] provides the cancer cell lines used, expression of activity (apoptosis, % inhibition,
and IC50), normal cell lines used, selectivity index, and IC50 of positive controls.
Table 1 Plant origin, plant parts used, and extraction methods used in previous studies.
Plant source (location)
|
Plant parts used
|
Extraction method
|
Ref
|
Commercial source 300 mg tablet plus inert excipients
|
Unknown
|
Tablet extracted with 2.2 mL 70% ethanol
|
[25]
|
Collected at Kirstenbosch (WCP), Free State (FSP), and Northern-Province (NP)
|
Leaves, stems, flowers
|
10 g of powder extracted in 1 L boiling water. 40 mg/mL stock solution
|
[26]
|
Gift from Dr. C. Albrecht, Medical Research Council (SA)
|
Leaves
|
1 g of dried leaves boiled in 10 mL water for 15 min
|
[27]
|
Murraysburg (SA) S. frutescens (L.) R.Br. var. microphylla
|
Leaves and small twigs
|
1 g extracted in 10 mL 70% ethanol for 24 h, stock solution (100 mg/mL)
|
[28]
|
Murraysburg (SA) S. frutescens (L.) R.Br. var. microphylla
|
Leaves and small twigs
|
0.5 g in 10 mL boiling water and left overnight
|
[29]
|
Two S. frutescens collected at Colesberg (A) and Platvlei (B) and S. tomentosa (C) collected at Stilbay
|
Leaves and stems
|
70% ethanol for 24 h and stock solutions in water (100 mg/mL)
|
[42]
|
S. frutescens subsp. microphylla
|
Leaves and twigs
|
0.5 g in 10 mL boiling water and left overnight
|
[30]
|
Location unknown–in ocean. Collected from same plant 1 year apart.
|
Leaves and small twigs
|
0.5 g in 10 mL boiling water and left for 15 h
|
[31]
|
PhytoNova (Cape Town, South Africa) in tablet form containing 300 mg of raw herb powder
compounded with inert excipients.
|
Unknown
|
Tablet extracted in 2.2 mL of 70% ethanol at room temperature for 2 h on a shaker.
Supernatant diluted 1/50 and 1/100
|
[32]
|
Big Tree Nutraceutical and Thebe Natural Medicines (Cape Town, South Africa)
|
Unknown
|
Extracted with methanol, dried, and redissolved in ethanol at 50 mg/mL
|
[33]
|
Sutherlandia SU1 tablets
|
Unknown
|
3 g S. frutescens in 30 mL of 70% ethanol, under constant agitation for 24 h.
|
[34]
|
S. frutescens R. BR. (variety Incana E. Mey.) purchased from Big Tree Health Products (Fish Hoek, South Africa)
|
Leaves
|
1 kg of the powdered preparation was soaked in boiling water and left overnight for
extraction.
|
[35]
|
Raw powdered plant material of S. frutescens purchased from Afrinatural holdings (KwaZulu-Natal, South Africa).
|
Unknown
|
1 : 10 ratio in water (plant : liquid)–10 g raw powder in 100 mL distilled water and
the suspension was sonicated at 45 °C for 45 min. A stock solution of 8 mg/mL was
prepared, and subsequent concentrations were prepared ranging from 1 to 8 mg/mL.
|
[37]
|
Collected from 7 locations throughout SA
|
Leaves
|
1 g leaves extracted with 20 mL of 75% ethanol. Ultra-sonicate for 60 min. Stock extract
of 1 mg/mL
|
[36]
|
Dried and milled, purchased from Afrinatural holdings
|
Unknown
|
1 : 10 ratio in water (plant: liquid) followed by 45 min sonication at 45 °C. 1 – 8 mg/mL
in culture medium stock solutions
|
[38]
|
Dried and milled, purchased from Afrinatural holdings
|
Unknown
|
1 : 10 ratio in water (plant: liquid) followed by 45 min sonication at 45 °C. 1 – 8 mg/mL
in culture medium stock solutions
|
[38]
|
Sutherlandia frutescens leaves purchased from Parceval Pharmaceutical Industry
|
Leaves
|
Macerated in MeOH (3 × 24 h) at room temperature
|
[19]
|
Table 2 Cell lines used, cytotoxicity, selectivity index, IC50, and positive controls of all studies testing S. frutescens.
nr
|
Cell lines used
|
[Apoptosis]
|
% inhibition
|
IC50
|
Normal cell lines (IC50)
|
Selectivity index
|
Positive control (IC50)
|
Ref
|
* Activity expressed as a dilution factor
|
1
|
MCF-7, MDA-MB-468, Jurkat, HL60
|
ND
|
ND
|
IC50 ± 1/250* (MCF7), 1/200 (MDA-MB-468), 1/150 (Jurkat) 1/200 (HL60)
|
ND
|
ND
|
Camptothecin 1 uM (348 ng/mL), paclitaxel 10 nM (8.54 ng/mL)
|
[25]
|
2
|
CHO, Caski, Jurkat T lymphoma
|
Observed at 3.5 mg/mL (WCP), > 10 mg/mL (FSP and NP)
|
ND
|
ND
|
ND
|
ND
|
ND
|
[26]
|
3
|
DU-145, MCF-7, MDA-MB-231
|
Minor inhibition at 50 ug/mL against DU-145 and MCF7 and stimulation of MCF-12A and
MDA-MB-231
|
ND
|
ND
|
MCF-12A
|
ND
|
Cisplatin 0.27 µg/ml (MCF-7), 0.14 µg/ml (MCF-12A)
|
[27]
|
4
|
MCF-7
|
70% ethanolic extracts induced morphological hallmark in MCF-7
|
40 – 50% inhibition after 72 h with 1.5 mg/mL plant material equivalents
|
|
ND
|
ND
|
No positive control (vehicle-treated controls)
|
[28]
|
5
|
MCF-7 and MCF-12A
|
ND
|
MCF-7 (26% at 10 mg/mL)
|
|
MCF-12A (49% at 10 mg/mL)
|
Claims selectivity but unclear both cell lines significantly inhibited–no IC50s provided
|
Unclear–no drug positive control. Vehicle controls
|
[29]
|
6
|
SNO
|
Extract A: 2.5 mg/mL: early apoptosis, 36% of cells and late apoptosis, 13% of cells. 5 mg/mL: early apoptosis 22% and late apoptosis 21%. Extract B: Early (25%) and late (64%) apoptosis at 2.5 mg/mL. Early (14%) and late
(74%) apoptosis at 5 mg/mL. Extract C: Early (30 – 32%) and late (31 – 32%) apoptosis at 2.5 – 5 mg/mL.
|
ND
|
ND
|
Cell viability reduction after 24 h: Extract A (2.5 and 5 mg/mL). B (0.5 – 10 mg/mL). C (1 – 10 mg/mL). After 48 h: Extract A (2.5 and 5 mg/mL). B (0.5 – 10 mg/mL). C (0.5 – 10 mg/mL).
|
ND
|
ND
|
[42]
|
7
|
MCF-7 and MCF-12A
|
ND
|
1 mg/mL at 48 h exposure GI50 parameters calculated for the MCF-7 (51.79% of control)
|
ND
|
MCF-12A (93.22% of control).
|
ND
|
No positive control (vehicle-treated controls)
|
[30]
|
8
|
MCF-7 and MCF-12A
|
MCF-7 (1/10, 20.59%),
|
ND
|
ND
|
MCF-12A (4.9% and 1.6%)
|
ND
|
Actinomycin D (0.1 µg/ml) and tamoxifen (20 µM)
|
[31]
|
9
|
CaCo2
|
Increased modulation of apoptosis-related proteins (PARP)
|
1/100 (no significant effect), 1/50 (48.83%)
|
ND
|
ND
|
ND
|
No positive control
|
[32]
|
10
|
Human (PC3 and LNCaP), Mouse (TRAMP-C2)
|
ND
|
ND
|
IC50 of 167 µg/mL in PC3, 200 µg/mL in LNCaP 100 µg/mL in TRAMP-C2
|
ND
|
100 µg/mL in TRAMP-C2
|
No drug controls
|
[33]
|
11
|
A375, Colo-800 melanoma; HDFα and Hek 293 normal cell lines
|
Stimulation of intrinsic and extrinsic caspase-dependent apoptosis
|
ND
|
A375 (625 µg/mL) but control cell more sensitive illustrating non-selectivity at these
high doses.
|
HDFα (300 µg/mL) Hek 293 (625 µg/mL 90% survival)
|
ND
|
Unknown
|
[34]
|
12
|
SKNBE(2) and SHSY5Y, KMST6
|
ND
|
ND
|
SKNBE (2) (2.3 mg/mL) and SHSY5Y (2.2 mg/mL) KMST6 at 5 mg/ml concentration cell survival was 50.9%.
|
ND
|
ND
|
ND
|
[35]
|
13
|
3D colorectal cell model (LS180)
|
ND
|
ND
|
LS180 (2.625 mg/mL)
|
ND
|
ND
|
Paclitaxel (94.595 nM)
|
[37]
|
14
|
DLD-1
|
ND
|
ND
|
4 samples > 200 µg/mL with Colesberg sample 158.7 ug/mL. No control
|
|
|
ND
|
[36]
|
15
|
LS180 cells and 2D cells
|
ND
|
ND
|
2630 µg/mL
|
ND
|
ND
|
Paclitaxel 0.081 µg/mL
|
[38]
|
16
|
LS180 cells 3D cells (spheroids)
|
|
|
Treated with 201 and 403 µg/mL
|
ND
|
ND
|
Treated with 0.006 µg/mL paclitaxel
|
[38]
|
17
|
MCF-7
|
ND
|
No 50% inhibition was achieved at 100 µg/mL for extract, fraction, and sutherlandiosides
and 1 saponin
|
ND
|
ND
|
ND
|
ND
|
[19]
|
The study conducted by Tai appears to be the first in vitro investigation on this topic and comprised the use of commercial Sutherlandia tablets [25]. A 70% ethanol extract used during this study was tested against the human mammary
adenocarcinoma (MCF-7; MDA-MB-468), human leukaemia (Jurkat), and human promyelocyte
(HL60) cell lines. The reported 50% inhibition (IC50) was expressed as a dilution factor of the primary 70% ethanol extract at the following
dilutions: 1/250 for MCF-7 (most active), 1/200 for MDA-MB-468, 1/200 for HL60, and
1/150 for Jurkat (least active). The positive controls, camptothecin and paclitaxel,
yielded IC50 values of 1 µM (348 ng/mL) and 10 µM (8.54 ng/mL), respectively.
The leaves, stems, and flowers of Sutherlandia were tested against Chinese hamster ovary (CHO), Caski (cervical carcinoma), and
Jurkat T lymphoma cell lines, where apoptosis was noted at 3.5 mg/mL for plants collected
in the Western Cape Province (WCP) and approximately 10 mg/mL for plants originating
from the Northern Province (NP) and Orange Free State (OFT) [26].
The cytotoxicity of an aqueous Sutherlandia extract was tested against prostate cancer (DU-145), breast cancer (MDA-MB-231 and
MCF-7), and non-malignant breast (MCF-12A) cell lines. The extract yielded IC50 values of > 50 µg/mL, which exceeded the limit of activity for crude extracts (30 µg/mL)
as set by the American National Cancer Institute. Interestingly, the positive control,
cisplatin, yielded respective IC50 values of 0.27 µg/mL (MCF-7) and 0.14 µg/mL (MCF-12A), indicating a lack of selectivity
[27].
Crude extracts of S. frutescens were tested for activity against the human breast adenocarcinoma (MCF-7) cell line,
and an IC50 value of 1.5 mg/mL was reported after 24 hours of exposure [28]. In a follow-up study of the cytotoxic effects of a crude S. frutescens extract on the MCF-7 and MCF-12A cell lines, a dose-dependent inhibition of 26% viable
cells (MCF-7) and 49% (MCF-12A) was reported at a concentration of 10 mg/mL compared
to a vehicle control. No positive drug controls were used in either of these studies
[28], [29]. In another follow-up study, an aqueous extract of S. frutescens against MCF-7 and MCF-12A was investigated. Inhibition of roughly 50% and 7% was
reported against the two cell lines, respectively, at a 1 mg/mL dose after 48 h exposure.
No positive drug controls were used [30].
An aqueous extract of S. frutescens was tested for activity against MCF-7 and MCF-12A cell lines, and optimal apoptosis
was shown for a 1/10 dilution. The MCF-7 cells revealed that 20.59% of cells treated
indicated apoptosis, whereas only 2.02% of MCF-12A cells indicated apoptosis [31]. In another study, the cytotoxic effects of S. frutescens on a colon cancer cell line (CaCo2) were investigated, and a 1/50 dilution of the
S. frutescens extract yielded a more significant decrease in cell viability compared to a 1/100
dilution [32].
The antiproliferative effects of S. frutescens in human (PC3 and LNCaP) and mouse (TRAMP-C2) cell lines were investigated, and IC50 values at 167 µg/mL, 200 µg/mL and 100 µg/mL against the PC3, LNCaP and TRAMP-C2
cell lines, respectively, were reported [33].
The apoptotic effects of S. frutescens in melanoma cell lines (A375 and Colo-800), as well as in HDFα and Hek 293 normal
cell lines, were tested. The results of this study yielded an IC50 value for the A375 cell line of 0.625 mg/mL following 24 h of treatment, whereas
the Colo-800 cell line showed higher resistance with 98% cell viability after 24 h
of treatment. The HDFα normal cell line showed high sensitivity to S. frutescens treatment with an IC50 value at 0.3 mg/mL. The Hek 293 normal cell line, however, showed the highest resistance
to treatment with a slight proliferative effect seen at a 0.625 mg/mL treatment concentration
between 24 and 48 hours of treatment [34].
The effects of S. frutescens were investigated on the cell viability of human malignant neuroblastoma cells (SKNBE(2)
and SHSY5Y) along with non-cancerous fibroblast cells (KMST6) using an MTT (3-[4,5-dimethylthiazol-2-yl]-2,5
diphenyltetrazolium bromide) cell viability assay and the clonogenic assay [35]. Moreover, flow cytometry was utilised to detect the accumulation of intracellular
reactive oxygen species (ROS), as well as the loss of mitochondrial membrane potential
(MMP). Caspase-9 activation was also used to detect cell apoptosis. IC50 values obtained were 2.3 mg/mL and 2.2 mg/mL for the SKNBE(2) and SHSY5Y, respectively,
whereas the KMST6 were less sensitive with an IC50 of 5 mg/mL.
The bioactivity of S. frutescens extracts from different regions via in vitro antioxidant and antiproliferative effects against the colon adenocarcinoma cell line
(DLD-1) was investigated [36]. The results of the antiproliferative assay revealed that all extracts produced
cytotoxic effects at the highest treatment concentration (200 µg/mL), and the most
potent antiproliferative activity was shown by the plant extracts originating from
Colesberg, Zastron, and Gansbaai 1 with respective IC50 values of 158.7 µg/mL, 172.7 µg/mL, and 176.7 µg/mL.
Two-dimensional and three-dimensional spheroid LS180 colorectal cancer cells were
used to test the activity of S. frutescens extracts. Commercial S. frutescens and Xysmalobium undulatum (L.) W. T.Aiton (Apocynaceae) were first tested against LS180 colorectal 2-D cancer
cells, and paclitaxel was used as positive control. IC50 values of 2.63 mg/mL and 0.098 mg/mL were reported for S. frutescens and X. undulatum, respectively, as compared to the positive control, which afforded an IC50 value of 0.081 µg/mL (S. frutescens were therefore 32,469 times less active than the positive control in 2D assays).
Half the IC50 concentration (1.315 mg/mL) of S. frutescens was used to test a newly established 3D spheroid model. They evaluated soluble protein
content, glucose consumption, intracellular ATP, and extracellular adenylate kinase
(AK) release and compared these parameters with the positive control,
paclitaxel. The authors conclude that “S. frutescens and X. undulatum crude aqueous extracts both showed notable cytotoxic effects and potential anticancer
activity relative to the known chemotherapeutic drug paclitaxel, with decreased growth,
cell viability, and glucose consumption following treatment” [37], [38].
The most recent study on Sutherlandia extracts was conducted by [19] who identified 8 new compounds and tested the extract, enriched fraction, and 10
of the 12 purified compounds against an MCF-7 cell line. No adequate inhibition at
the highest concentration of 100 µg/mL tested was observed.
Critical Evaluation
There are several minor but also major concerns that need to be discussed. The minor
concerns will first be detailed, followed by a more in-depth discussion of the major
concerns. The study conducted by [25] does not provide a precise IC50 value ([Table 2]), even though it is relatively uncomplicated to determine. However, when the IC50 value of the positive control (8.54 ng/mL) is considered and an extraction yield
of 10% is assumed, then the IC50 value against MCF-7 can be calculated as 54 µg/mL (roughly 6300 times less active
than the positive control). This is an estimate with the authors stating that the
IC50 “is about 1/250′′ dilution. Moreover, for the MCF-7 cell line, the positive control
as indicated in Fig. 3 of [25] clearly inhibits the cells far below its IC50 value with only about 10% of cells surviving at the concentration
tested. The IC50 is therefore likely to be significantly lower than reported and, subsequently, the
difference between the S. frutescens sample and positive control much larger than reported.
In the study conducted by [26], it is unclear where the plant material was collected ([Table 1]). It mentions Kirstenbosch, Western Cape Province, “Bloiemfontent”, “Free State”,
and Northern Province, but it is not specified what these locations refer to as “Bloemfontein”
is the capital city of the Free State province in South Africa and no Northern Province
exist (the North-West and Northern Cape provinces do exist). Apoptosis was observed
in one sample at a concentration of 3.5 mg/mL, but no apoptosis was detected in the
other two samples ([Table 2]). No positive control or non-cancerous cell lines were used, and hence, no selectivity
index was determined. The conclusion states: “In summary, the claim by traditional
healers that Sutherlandia frutescens has anticancer properties has been partially validated by identifying an extract
in the plant that has marked apoptotic activity”.
We tend to disagree with this conclusion when the extremely high concentrations tested
(3.5 mg/mL) are considered.
The findings highlighted by the study conducted by [27] do not specify the source of the plant material, only stating that it was a gift
from the Medical Research Council of South Africa. The study does not provide a specific
IC50 value (rather % inhibition), and S. frutescens performed poorly compared to five other plant species tested. The authors included
positive controls and used acceptable guidelines to conclude that S. frutescens did not show acceptable activity and therefore continued work on other plant species.
The source of plant material was clearly defined by the study conducted by [28]; however, the study did not include positive controls or non-cancerous cell lines.
They expressed the IC50 as 1.5 mg dry plant material/mL; therefore, no exact IC50 was provided. Given an extraction efficiency of 10%, the actual IC50 is likely around 10 times lower. In a very similar study, [29] once again clearly defined their source of plant material but prepared an aqueous
extract instead of a 70% ethanol extract. Again, no positive controls were used but
a non-cancerous MCF-12A was included. At a high concentration of 10 mg/mL, some selectivity
was claimed with the plant extract leading to a survival of only 25% of the MCF-7
cells vs. 54% of the MCF-12A cell line. However, from Fig. 1 in [29], it is clear that the reverse seems to occur at 5 mg/mL, with an estimated 64%
survival of MCF-7 vs. 56% against MCF-12A. The paper states: “The non-tumorigenic
MCF-12A cell line is less susceptible to apoptosis induction at higher concentrations
of S. frutescens compared to the tumorigenic MCF-7 cell line.” The observed selectivity at 10 mg/mL
vs. 5 mg/mL is, in our view, more a function of the high dosage, which will kill the
cell (and will likely kill any cell type) with the slight ‘selectivity’ only observed
under certain conditions due to inherent differences in cell types used–therefore,
no true selectivity.
In a follow-up study conducted by [30], the same cell lines were used (MCF-7 and MCF-12A), and a dose of 1 mg/mL and 48 h
of exposure was chosen based on a dose-response study. The authors do show that by
using these dosing parameters, a certain level of selectivity is evident with the
MCF-7 being more sensitive. It is, however, clear from Fig. 1 in [30] that, at different concentrations or exposure times, the exact opposite seems to
occur with the MCF-12A being more sensitive than the MCF-7 cell line (e.g., at 2.5 mg/mL).
Moreover, no positive controls were included, making these results hard to interpret.
In [Table 1] of [31], the source of the plant material is not clearly specified with the coordinates
provided (35°51′32.12″S; 18°39′45.69″E) pointing to a location in the ocean. For sample
preparation, the study used 10 mg/mL of plant material in water (rather than an extract
concentration). Fig. 2 in [31] is unclear, making it difficult to understand how the differences could be considered
“significant”. The significance is expressed compared to the growth medium (vehicle)
control and does not include any statistically significant differences between the
MCF-7 and MCF-12A cell lines. The manuscript mainly deals with the “differential cellular
interaction” and in essence indicates that there may be a different mode of action
against the two cell lines (apoptosis vs. autophagy) but no major difference in selectivity.
In the study conducted by [32], dilutions are utilised instead of specifying exact concentrations. The study lacks
a positive control and does not provide an IC50 value. The 1/100 dilution did not provide any significant activity, while the 1/50
dilution inhibited the Caco2 cells (48.83% survival). A 1/50 dilution equates to roughly
272 µg/mL when a 10% extraction yield is used in the calculations.
The study carried out by [33] included both in vitro and in vivo tests (although it lacked positive controls, and the plant parts used in this study
remain unknown). The study also tested a fraction, mainly consisting of sutherlandioside
D, and reported an IC50 value of 1.8 µg/mL compared to 30 µg/mL for the crude extract against stimulated
Shh Light II cells. It is unfortunate that sutherlandioside D was not also tested
against the PC3, LNCaP, and TRAMP-C2 cell lines, making comparison of the results
difficult. The in vivo mice experiments yielded some unforeseen results. Sutherlandia frutescens was mixed with mouse diets at three concentrations (0.05%, 0.25%, or 1% (wt/wt))
and fed to mice who developed autochthonous prostate tumours. No dose response could
be observed, as with the in vitro experiments, and the 0.05% yielded the best results. However, it is unclear if the
difference between the 0.05%
treatment group and the control group is statistically significant (Fig. 7 in [33]–data not provided).
No positive controls were included in [34], and they reported a 50% inhibition at 625 µg/mL. However, the non-cancerous cell
line HDFα afforded a roughly 50% inhibition at 300 µg/mL, indicating a lack of selectivity.
The authors correctly concluded, “However, the relatively high concentration of the
extract required to elicit such a response may pose a significant limitation for the
clinical use of S. frutescens for the treatment of cancer.”
No positive controls were included in the study conducted by [35]. They do, however, show a selectivity index of roughly 2 between the SKNBE and KMST6
cell lines but at extremely high dosages (IC50 of 2.3 vs. 5.0 mg/mL). One study was not included in the tables due to the difficulties
in interpreting the results [39]. Sutherlandia frutescens, canavanine, GABA, and pinitol were tested for activity against SiHa cervical cells.
From Fig. 1 in [39], it is abundantly clear that absolutely no dose-response curve can be observed.
It is entirely unclear from the data presented how IC50 values for all samples tested could be calculated. For the S. frutescens extracts tested at 50, 100, and 200 µg/mL, all concentrations afforded ~ 50% cell
survival, whereas at 150 µg/mL, cell viability was presented as 250%. The same trend
can be observed for the three compounds,
with the same inhibition pattern observed at all concentrations tested.
Sutherlandia frutescens samples were collected from seven different geographic locations and tested against
DLD-1 colon cancer cells [36]. Unfortunately, no positive or vehicle controls were used. The best activity of
158.7 µg/mL was reported for one specific sample with four samples presenting IC50 values of > 200 µg/mL. This indicated that chemotypic variation may affect the bioactivity,
but in the absence of controls, it remains difficult to form any firm conclusions.
The findings reported by [38] reported activity of S. frutescens extracts that are ~ 32 500 times less active than the positive control in 2D assays.
The positive control paclitaxel afforded an IC50 value of 0.081 µg/mL, and if this is extrapolated with an average chemotherapeutic
dose of 135 and 175 mg/m² for paclitaxel, it will equate to an S. frutescens dose of 4.39 – 5.69 kg/m2, which is clinically
irrelevant. However, the study concludes that “the results of this study suggest that
the anticancer activity of the S. frutescens crude aqueous extract, specifically in colon cancer, may even surpass that of the
model chemotherapeutic drug, paclitaxel”. We simply disagree with this statement,
and this brings us to a discussion of the major issues in many of the publications
that need to be highlighted.
There are three main and, in our view, serious concerns. First, the absence of proper
controls, especially positive controls. Positive controls are crucial for several
reasons. First, bioassays involving living cells or organisms, both in vitro and in vivo, can be challenging and prone to significant variability in pharmacological response.
Thus, incorporating a positive control is essential to confirm that the test system
is functioning correctly. A negative control, such as a solvent or carrier, can also
fulfil this role to some degree, and ideally, both a positive and negative control
should be used [40]. Second, a positive control provides a benchmark for comparison, allowing researchers
to evaluate whether results, such as the IC50 of the positive control, are consistent with previous findings or literature and
how the test sample compares to the positive control. Many academic journals (such
as the Journal of
Ethnopharmacology, Phytomedicine Plus, Planta Medica, etc.) emphasise the importance of positive controls, and it is stated in the author
guidelines that the absence of the use of proper controls (e.g., recognised positive
controls) may lead to immediate rejection of a manuscript [41]. This policy underscores the essential role that positive controls play in maintaining
the integrity of scientific research. Without a positive control, it becomes impossible
to validate the assayʼs performance, raising doubts about the accuracy of the results
and the robustness of the experimental design [42]. Furthermore, positive controls provide a frame of reference that ensures consistency
across scientific studies. For example, researchers can compare the activity of a
test sample (such as S. frutescens) to that of a well-established active drug (such as paclitaxel), facilitating meaningful
comparison and interpretations of
the findings. By adhering to this standard, researchers not only increase the credibility
of their work but also contribute to the overall advancement of science, preventing
the publication of misleading or inconclusive data.
Only five of the studies ([Table 2]) made use of positive controls [25], [27], [31], [37], [38]. However, many studies failed to include positive controls or only used solvents
as controls, making their findings far less useful and reliable [26], [28], [29], [30], [32], [33], [34], [35], [36], [43]. It was stated that “If no positive control was used in a standard assay, it is
impossible to interpret the resulting data with regard to their scientific content”
[44]. The importance of positive
controls can also be explained by precision and accuracy. An increase in sample replicates
will lead to a more precise result but cannot provide accuracy. Only a positive control
can provide accuracy; hence, the lack of a positive control may lead to very precise
but completely inaccurate results.
The other main concerns are the exceptionally high doses tested and the lack of selectivity
studies, which, in essence, is the holy grail of drug discovery in the field of cancer
research. Regarding high doses, the National Cancer Institute of the United States
uses the following guidelines regarding plant extracts: IC50 < 10 µg/mL, highly active; 10 – 50 µg/mL, moderately active; 50 – 200 µg/mL, weak
activity; and > 200 µg/mL, inactive [45]. It was also suggested that an IC50 < 100 µg/mL for extracts and < 25 µM for compounds should be considered the minimum
acceptable level of activity [46], while Gertsch recommends < 50 µg/mL and < 5 µM, respectively [47]. Here, Paracelsus’ dictum, “The dose makes the poison”, which emphasises the importance
of dose in determining efficacy and safety, can be highlighted [46], [47].
As an example, the study done by [29] can be discussed. Other than the lack of a positive control, the dosage used was
10 and 5 mg/mL (at a 10% extraction yield, it will equate to a dosage of 1.0 and 0.5 mg/mL).
These high doses will likely kill any cell type, which was eloquently illustrated
by the different responses observed between the cancerous MCF-7 and non-cancerous
MCF-12A at 10 mg/mL, where MCF-7 was more susceptible, but at 5 mg/mL, the MCF-12A
was more susceptible. Moreover, even if an IC50 of 1 mg/mL is assumed and this value is compared to a positive control, paclitaxel,
as published by [25] against MCF-7 of 8.54 ng/mL, a ~ 120 000-fold difference in IC50 values is observed. When this is translated into an in vivo chemotherapeutic dose (135 – 170 mg/m2 for paclitaxel) many kilograms/m2 of S. frutescens must be administered, which is simply not
possible. Other high-dose experiments, such as [26] who reported inhibition at 3.5 mg/mL, [43] at concentrations of 2.5 – 5 mg/mL, [35] who tested at 2.3 mg/mL, [37] at 2.625 mg/mL, and [38] at doses of 2.63 mg/mL, are highly concerning, as these high doses are clinically
irrelevant. These have been called “outrageously high concentrations”, adding that
“…claims of activities at absurd concentrations are extrapolated to the human situation…”
[44]. We tend to agree and must conclude that most of the studies presented here were
tested at far too high concentrations to make the studies scientifically meaningful.
The selectivity index (SI) is another crucial aspect and is described as the ratio
between the toxic concentration of a sample relative to its bioactive concentration.
Furthermore, an ideal drug or active compound would possess a high toxic concentration
directed towards malignant cells while maintaining a low bioactive concentration [48], [49]. Determining the SI during any research surrounding herbal medicine or isolated
active compounds is necessary to evaluate if further studies are warranted [49]. Several studies investigated the selectivity of S. frutescens extracts between the tumorigenic breast cancer cell line MCF-7 and the non-tumorigenic
MCF-12A cell line [27], [29], [30], [31]. The ideal situation would be that the MCF-7 cell line would be highly sensitive
to
S. frutescens, whereas the MCF-12A cell line would not be sensitive at all, resulting in a high
SI. In essence, the higher the SI, the better. Unfortunately, only two studies included
the positive controls, cisplatin [27] and tamoxifen [31], which places a question mark on the validity of the other studies. Moreover, excessively
high doses were selectively tested, and in addition, no SI values were presented even
though the study designs are essentially focused on calculating an SI value. It was
shown that at 50 µg/mL, little to no activity was observed against the MCF-7 cell
line, whereas some stimulation was observed in the MCF-12A cell line [27]. It is of interest to note that the positive control also did not show any selectivity
as the IC50 of cisplatin was given as 0.27 µg/mL and 0.14 µg/mL against the MCF-7 and MCF-12A
cell lines, respectively. Due to the low activity
of S. frutescens, no IC50 value was provided and, hence, no SI was presented with the authors correctly discontinuing
further work on S. frutescens based on the observed data.
The data presented by [29] was difficult to interpret. The MCF-12A cell line seems to be more sensitive to
S. frutescens extracts at 3 and 5 mg/mL, whereas the MCF-7 cell line is slightly more sensitive
at 10 mg/mL. Based on this, a decision was made to continue more advanced studies
at the higher dosages, such as flow cytometry and electron microscopy, whilst the
decision should have been that there was no clear selectivity at these very high dosages,
and the focus and resources should have been spent on more promising candidates.
In the study by [30], the same pattern emerges. At very specific (high) concentrations and treatment
times, MCF-7 seems to be more sensitive, but at different concentrations and treatment
times, the opposite appears to happen. Here again, more advanced studies were conducted
at a very specific concentration and exposure time, where there seems to be a slight
difference in favour of MCF-7, whereas an unbiased opinion would have been that the
quality of data was not good enough, and scarce resources should have been spent on
more promising candidates.
Of the 17 studies in [Table 2], only one discussed an SI [33]. The remaining studies did not mention any calculated SI value even though they
alluded to the presence of selectivity. Evaluating the antiproliferative activity
of a sample using only malignant cell lines, without considering the SI, provides
a poor indication for further clinical studies [50]. Optimally, with the provision of an SI, a drug should be able to inhibit cancer
cells at a very low concentration while normal cells should not be affected during
in vitro scientific studies [51]. Hence, without the SI data for the reported bioactive samples, the value of such
publications is significantly limited [52].
Limitations of In Vitro Models
Limitations of In Vitro Models
In vitro models are used because they are relatively inexpensive and easy to perform and many
samples can be tested simultaneously, making it an ideal tool in large drug-discovery
projects. Moreover, there are fewer ethical concerns with in vitro studies than with in vivo animal studies. It essentially acts as a funnel to filter out the inactive or less
active samples in order to focus on the most promising leads, which can then be further
studied in much more expensive in vivo models [53]. It is essential to recognise that laboratory-based cytotoxicity tests (in vitro) do not consider important pharmacokinetic factors like absorption, metabolism, and
excretion, which are critical in actual clinical trials (in vivo) [54]. Consequently, the outcomes of in vitro cytotoxicity studies should be viewed as an approximate guideline rather than direct
indicators of in vivo
cytotoxicity [54]. The old adage “In vitro simplicitas, in vivo veritas” (in vitro simplicity, in vivo the truth), as stated by [44], should here be considered. It is therefore unfortunate that no in vivo studies, other than the small-scale mice experiments [33], have been conducted, and yet major claims regarding the anticancer activity of
S. frutescens are made based on in vitro results, most of which tested at (extremely) high doses and excluded the use of proper
positive controls or selectivity studies.
Future Research Recommendations
Future Research Recommendations
Regarding S. frutescens, the availability of in vivo data pertaining to its antiproliferative/anticancer activity appears to be lacking.
In vivo studies are essential to determine the actual effectiveness of Sutherlandia and whether it can be safely and effectively incorporated into cancer treatment regimens.
Only two documented clinical trials could be found, and it is important to note that
they were to test tolerability [23] and were on HIV patients and not on cancer patients [12].
This literature review may be construed as overly critical or even negative, but in
our view, the general poor quality of published research lends itself to the needed
critique. Using established guidelines, current research indicates minimal to no in vitro activity of S. frutescens against the tested cancer cell lines. However, a common counterargument, driven by
extensive anecdotal evidence and the herbʼs popularity, suggests potential in vivo activity, possibly due to active compounds being present at very low concentrations
that standard bioassays will fail to detect. This hypothesis is plausible, given the
well-documented chemical complexity of plant extracts, which can mask bioactivity
through unwanted cellular interactions [55]. To conduct in-depth research on S. frutescence, techniques to reduce chemical complexity, as was published by the National Cancer
Institute, which developed a rapid prefractionation
procedure before in vitro bioactivity testing, should be employed [56]. Another explanation that is often provided for the lack of in vitro activity is that potential active compounds may exist as inactive prodrugs in the
extract, undetectable in standard in vitro assays. Techniques to activate prodrugs for in vitro detection include incubation with human intestinal microbiota [57], enzymatic hydrolysis with β-glucosidase [58], incubation with liver microsomes [59], and various chemical hydrolysis techniques [60].
To guide future research on S. frutescens, the following stepwise approach is recommended:
-
Collect specimens from as many sources as possible to create sufficient chemical diversity.
-
Perform chemical fingerprinting (e.g., metabolomics) and in vitro bioactivity testing.
-
Prefractionate the most promising samples and repeat step 2.
-
If no significant bioactivity is detected, apply metabolic activation techniques and
retest.
-
If these steps fail, consider in vivo animal studies or clinical trials, though ethical justification may be challenging,
and hence, it must be concluded that S. frutescens lacks significant activity.
For well-characterised medicinal plants such as Cannabis sativa L. (Cannabaceae) containing the active cannabinoids, Artemisia annua L. (Asteraceae) containing artemisinin, or Mesembryanthemum tortuosum L. (Aizoaceae) containing mesembrine alkaloids, the known active compounds enable
quality control and production of standardised preparations. In contrast, the unknown
active compounds in S. frutescens preclude effective quality control, making the identification of active compounds,
if present, a critical priority.
Moreover, the gap between in vitro and in vivo studies during medicinal plant drug discovery, due to the limitations of traditional
in vitro models in replicating complex physiological conditions, hinders the progress of developing
new therapies. Advanced in vitro models, such as organoids, organ-on-a-chip systems, and 3D bioprinted tissues, can
accurately mimic human tissues, enabling precise analysis of plant compounds for therapeutic
efficacy and potential toxicity [61], [62], [63]. Computational tools, including PK/PD modelling and even artificial intelligence
platforms, can predict in vivo behaviour from in vitro data through analysis of complex datasets, correlating in vitro target inhibition with in vivo cell activity, which is particularly useful for identifying and evaluating plant-derived
compounds. These models may mediate
the translatability of in vitro results to in vivo outcomes by identifying patterns and predicting possible drug-target interactions
[64], [65]. Ex vivo tissue models validate in vitro results, assessing compound absorption and metabolism [66]. Microphysiological systems and biomarker-driven assays enhance physiological relevance,
while integrated platforms combining traditional knowledge and omics data streamline
drug discovery [67], [68], [69]. Nanotechnology can address challenges like low solubility, thus improving translatability
[64]. A phased approach, such as adopting advanced models, scaling multi-organ systems,
and standardising platforms, could potentially enhance scientific research into S. frutescens and subsequently may lead to
the development of plant-derived therapies.
Conclusions
Based on the available literature, we must conclude that research conducted to date
on the antiproliferative effects of Sutherlandia is mainly promotional, with a few exceptions, such as that published by [27]. The level of obfuscation that was found in many publications is a serious cause
of concern. Considering the serious nature of cancer, we find this to be unconscionable
and alarming, and this negatively impacts this research field. Conclusions such as
“partially validated” and “…anticancer activity of the S. frutescens … may even surpass that of the model chemotherapeutic drug, paclitaxel” are misleading,
to say the least, considering the major limitations and shortcomings of almost all
published studies. A simple rule of thumb is not to make any clinical claims based
on in vitro data alone. A striking illustration of our ‘promotional research’ statement is how
a negative report was reframed positively by referencing an
early mention of Sutherlandiaʼs use nearly 100 years later. In the original text,
as published in 1895, and somewhat typical of the time, it is stated that “There can
be no reasonable doubt that Sutherlandia has been the means of curing malignant tumours, cancerous in appearance, which were firmly believed to be cancers by non-professional persons unacquainted
with the distinctive marks of typical cancer. It is also certain, that employed as blood purifier and tonic, it has delayed the progress of true cancer and much prolonged life. In view of these facts and of the confident assertions made regarding Sutherlandia, it was desirable that it should be tested in a typical case of cancer, and that
was done recently by a physician of high intelligence with the result, as in some
similar instances formerly, that it proved wanting though applied both externally and internally” (boldface and underlining added) [5].
This paragraph was ‘cited verbatim’ from the original text but was presented as “curing
of malignant tumours, cancerous in appearance; also used as blood purifier and tonic
… to delay the progress of true cancer and much prolonged life” [4]. That it was tested and found wanting was simply omitted, and this is, in our view,
promotional research.
Based on the available information and keeping in mind possible herb-drug interactions
[70] and the serious nature of cancer, which causes over 10 million deaths annually,
S. frutescens should currently not be recommended for use. To answer the question being asked in
the title: currently, there seems to be no scientific merit in the anticancer claims
made for S. frutescens, and research to date is mainly promotional.
Contributors’ Statement
CRediT authorship contribution statement Nicezelle Gernandt: writing – original draft,
review and editing. Frank Van der Kooy: conceptualisation, supervision, data curation,
writing – original draft, review and editing.