You are on page 1of 7

Journal of Ethnopharmacology 277 (2021) 114105

Contents lists available at ScienceDirect

Journal of Ethnopharmacology
journal homepage: www.elsevier.com/locate/jethpharm

Suppressive, curative, and prophylactic potentials of an antimalarial


polyherbal mixture and its individual components in Plasmodium
berghei-Infected mice
Stephenie C. Alaribe a, *, Akolade R. Oladipupo a, Goodness C. Uche a, Maryan U. Onumba a,
Duncan Ota d, Olufunsho Awodele b, Wellington A. Oyibo c
a
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, College of Medicine Campus, PMB 12003, Idi-araba, Lagos, Nigeria
b
Department of Pharmacology, Therapeutics & Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
c
Department of Medical Microbiology & Parasitology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
d
Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria

A R T I C L E I N F O A B S T R A C T

Keywords: Ethnopharmacological relevance: Malaria remains one of the most prevalent infectious diseases in tropical regions
Antiplasmodial activity of the world, particularly in sub-Saharan Africa, where it remains epidemiologically holoendemic. The absence of
Malaria chemosuppression effective vaccines and Plasmodium resistance to antimalarial drugs have been the major challenges to malaria
Malaria parasite clearance
control measures. An alternative strategy could be the application of validated and standardized herbal
Polyherbal decoction
Plasmodium berghei
formulations.
Aim of the study: To evaluate the antimalarial activity of a polyherbal mixture (APM) and compare it to those of
its individual constituent plants.
Methods: APM consisted of stem barks of Mangifera indica (MI), Azadirachta indica (AI), Nauclea latifolia (and
roots, NL) and roots of Morinda lucida (ML). Dihydroartemisinin-piperaquine (DHP) and pyronaridine-
artesunate (PA) served as positive controls. Antimalarial activity was evaluated using suppressive, curative
and prophylactic assays in mice infected with Plasmodium berghei.
Results: All the herbal mixtures, individually and in combination, showed significant (p < 0.05) antiplasmodial
activities in the various assays. They produced considerable parasite suppression (>50%), substantial clearance
(>70%), and notable prophylaxis (>60%, except for NL: 35%). APM (95.4–98.7%) and AI (92%), respectively,
elicited greater and comparable suppression relative to DHP (88%) and PA (87.3%). However, all the herbal
decoctions, individually (72–93.6%) and in combination (82.5–91%), showed lower parasite clearance than DHP
(100%) and PA (99.5%). Meanwhile, APM showed relatively greater suppression and prophylaxis than its
constituent plants, suggesting that the combination produced synergistic or additive effects.
Conclusion: These findings could substantiate the use of these plants, singly or in combination, as traditional
remedies for malaria. Further studies are recommended to evaluate their clinical usefulness.

1. Introduction concentrated in 13 countries, and over half in Nigeria, Congo, Ethiopia,


Tanzania and Kenya. Furthermore, recent World Malaria Report indi­
Malaria is one of the most virulent of the recognized infectious dis­ cated that malaria episodes and mortalities have decreased by approx­
eases affecting man. This disease continues to wreak havoc to human imately 8% and 28%, respectively from 2010 to 2017 (WHO, 2018). This
beings especially in tropical and subtropical regions—particularly in suggests that some progress has been made in malarial control in this
sub-Saharan Africa, where it remains epidemiologically holoendemic past decade, which could be attributed to the different chemothera­
(Alaribe et al., 2020). World Health Organization (WHO) (2018) esti­ peutic and non-chemotherapeutic interventions. However, the latest
mated that 1.2 billion people are at high risk of malaria transmission, data also shows that there are still significant works to be done,
half of which live in the African regions; 80% of such cases are particularly if we are going to get anywhere close to meeting the WHO’s

* Corresponding author.
E-mail address: salaribe@unilag.edu.ng (S.C. Alaribe).

https://doi.org/10.1016/j.jep.2021.114105
Received 15 December 2020; Received in revised form 20 March 2021; Accepted 3 April 2021
Available online 5 May 2021
0378-8741/© 2021 Elsevier B.V. All rights reserved.
S.C. Alaribe et al. Journal of Ethnopharmacology 277 (2021) 114105

(2015) ambitious target of “reducing the global malaria burden by 90% mill. The decoction of each component was prepared by boiling 100 g of
by 2030”. The major challenges jeopardizing efforts in this regard are pulverized material in 1 L of distilled water for 24 h. For NL, 50 g each of
the rapid development and spread of Plasmodium resistance to existing both stem barks and roots were used. For APM, the aforementioned
and emerging antimalarial drugs on the one hand and mosquito resis­ quantities of the different plant components were mixed together in the
tance to insecticides on the other hand. It has been widely articulated ratio 1: 1: 1:1 and boiled in 4 L of distilled water for 24 h. The collected
that the best long-term control option is the development of vaccines, liquid extracts were lyophilized and stored in a refrigerator at -20 ◦ C
which could effectively prevent the spread of the disease. Nevertheless, until used.
all the promising vaccines developed so far are still undergoing clinical
trials (Bejon et al., 2008; Guinovart et al., 2009; Abdulla et al., 2013; 2.2. Experimental animals and malaria parasite
Moreno and Joyner, 2015; Butler and Stricker, 2019). Another approach
in malaria control is the application of traditional medicines, particu­ Swiss albino mice (weighing 20–25 g; 4 weeks old) of either sex were
larly plant-based remedies for the treatment of malaria. In fact, this obtained from Animal Facility Centre of College of Medicine, University
practice is as old as malaria itself. Moreover, some of the current anti­ of Lagos, Nigeria and maintained under standard environmental con­
malarial drugs, including amodiaquine and artemisinin, were either ditions of temperature (≈25 ◦ C), relative humidity (≈50%) and illumi­
isolated from plants or developed using plant’s isolates as lead molecules nation (≈12/12 h light/dark cycle). The animals were provided with
(Alaribe et al., 2020). Thus, it seems logical to continue to investigate standard pellet feed and drinking water and allowed to acclimatize for 7
plants and other natural products, used traditionally for malaria treat­ days prior to the commencement of the study. Animals were used in
ment, as potential source of novel antimalarial candidates. However, accordance with the internationally accepted ‘Guide for the care and use
since these natural products are traditionally often applied as of laboratory animals’ (National Research Council, 1996). The study was
multi-component formulations, it is also important to investigate their conducted with approval from the Health Research Ethics Committee of
therapeutic properties in their traditional forms. In view of the forego­ College of Medicine, University of Lagos with document Number;
ing, the attention that is accorded the conventional bioactivity-guided CMUL/HREC/03/18/341.
isolation of individual molecules should be extended to validation and Chloroquine-sensitive Plasmodium berghei (NK65 strain) was ob­
standardization of multi-component herbal formulations for acceptance tained from the National Institute of Medical Research (NIMR), Lagos
for clinical use, particularly in malaria management. It could be spec­ and was maintained by serial passage in different donor mice.
ulated that owing to the complexity of these multi-component herbal
formulations, it may be unlikely that Plasmodium will develop resistance 2.3. Parasite inoculation
to these multi-component herbal formulations, which has been the bane
of orthodox antimalarial drugs. Moreover, studies have shown that some Parasite inoculation was done as previously described (Fidock et al.,
crude plant extracts exhibited greater antiplasmodial activity than in­ 2004). Briefly, mice previously infected with P. berghei (NK65 strain)
dividual antimalarial drugs, such as artemisinin, at an equivalent dose having a parasite load of 20–30% were used as donor. Blood samples
(Wan et al., 1992; Wright et al., 2010; Rasoanaivo et al., 2011). were collected from the donor mice through ocular puncture into hep­
A large number of medicinal plants are used for treating malaria in arinized vacutainer tube. Subsequently, the blood was diluted with
Nigeria, particularly in the Southern regions of the country, where rain physiological saline (0.9%) in such a way that 1 ml of the final sus­
forests and humid tropical climate exist, which are ideal conditions for pension contained 5 × 107 P. berghei-infected red blood cells (RBCs). All
persistent malaria transmission all year round (Adebayo and Krettli, the mice used for the antiplasmodial investigation were inoculated
2011). One out of the many groups of herbal remedies used in Nigeria is intraperitoneally with 0.2 ml of infected blood (equivalent to 1 × 107
the antimalarial decoctions popularly called Agbo iba, which receive P. berghei-parasitized erythrocytes).
great patronage, majorly in the Southern regions of the country (Alaribe
et al., 2018). The composition, method of preparation and dosage of 2.4. Grouping and dosing of animals
these antimalarial decoctions vary from vendor to vendor as earlier re­
ported (Alaribe et al., 2018), but usually, these decoctions are often The animals were grouped and dosed for the antiplasmodial inves­
polyherbal, prepared in water and sometimes alcohol, which serves as tigation as follows. For both suppressive and curative tests, separately,
preservative and also as desired by vendors and consumers. In this study, 50 mice were randomly grouped into 10 groups (I–X) of 5 mice each.
we comparatively evaluated the antimalarial activities of a polyherbal Groups I, II and III were orally administered APM at 100, 160 and 320
mixture and individual plant of the polyherbal mixture with the stan­ mg/kg/day (i.e., 0.162, 0.259 and 0.518 mg/kg human equivalent doses
dard antimalarial drugs on mice infected with chloroquine-sensitive (HED)), respectively. Groups IV, V, VI and VII were administered 100,
Plasmodium berghei (NK65 strain). The antimalarial polyherbal mixture 1000, 1200 and 1240 mg/kg/day (i.e., 0.162, 1.62, 1.94 and 2.0 mg/kg
(APM) consists of the stem barks of Mangifera indica (MI), Azadirachta HED) of NL, MI, ML and AI, respectively, based on their active doses and
indica (AI), and Nauclea latifolia (and roots, NL) as well as roots of LD50 values as previously reported (Okpanyi and Ezeukwu, 1981; Awe
Morinda lucida (ML). These plants have been reported to be used for the et al., 1998; Usha Devi et al., 2001; Taïwe et al., 2011; Umar et al., 2013;
treatment of different diseases, including malaria (Awe et al., 1998; Adejo and Atawodi, 2014; Chidozie et al., 2014; Kouadio et al., 2014;
Usha Devi et al., 2001; Umar et al., 2013; Alaribe et al., 2020). Ettebong et al., 2015; Alaribe et al., 2020). Group VIII, negative control,
received distilled water, while groups IX and X, positive controls, were
2. Materials and methods administered 2:18 and 4.5:1.5 mg/kg/day (i.e., 0.0032:0.0292 and
0.0073:0.0024 mg/kg HED) of dihydroartemisinin-piperaquine (DHP)
2.1. Collection and preparation of plant materials and pyronaridine-artesunate (PA), respectively. For the prophylactic
test, 40 mice were used and grouped into 8 groups (I–VIII) as described
The different plant materials: MI, AI, NL and ML were purchased above. The HEDs were calculated according to USFDA (2005)
freshly from Mushin herbal market, Lagos, Nigeria. The plant materials guidelines.
were identified and authenticated by a taxonomist, Dr. G.I. Nodza at the
Herbarium of the Department of Botany, Faculty of Science, University 2.5. Test for suppressive activity (Peter’s 4-day test)
of Lagos, Lagos, Lagos state. The herbarium specimens were deposited
and the following voucher numbers assigned: MI (LUH 8619), AI (LUH Suppressive activities of APM and its components: MI, AI, NL and ML
8618), NL (LUH 8622) and ML (LUH 8621). The plant materials were air were evaluated on early P. berghei infection in mice as described by
dried at room temperature and ground to coarse powder using an impact Knight and Peters (1980). The mice were inoculated, grouped and dosed

2
S.C. Alaribe et al. Journal of Ethnopharmacology 277 (2021) 114105

as described in section 2.4. The different treatments were started 3 h


after infection and were administered as a single dose per day for four
consecutive days (days 0–3). On the fifth day (day 4 i.e., 96 h
post-infection), blood samples were collected from tail snips of the mice.
Thick and thin films were made on the same slide for each blood sample.
The thin films were fixed in methanol and air dried. The blood films
were stained with 3% Giemsa at a pH of 7.2 and examined under a light
microscope using × 100 objective lens (immersion oil). The number of
malaria parasites and leucocytes per high power field (HPF) were
counted and parasite density was determined, assuming an average
leucocyte count of 8000 per μl, using the following equation (Green­
wood and Armstrong, 1991).

Parasite density (No. of parasites/μl) = (No. of Parasites/No. of leucocytes


counted) x 8000 (1)

A slide was considered negative after 100 HPF have been examined.
Average percentage chemosuppression (or parasite clearance) of each
treatment was calculated from the average parasite density of the
Fig. 1. Suppressive activity of antimalarial decoctions and controls against
negative control group (A) and the average parasite density of the test
P. berghei infection in mice. Each bar represents the mean ± SEM; n = 5. Values
group (B) using the following expression.
in each group are significantly different (p < 0.05) from the other groups.
Average percentage (%) chemosuppression = [(A – B)/A] x 100 (2)
suppressed parasitaemia than each of its components and the positive
controls. NL (100 mg/kg), MI (1000 mg/kg) and ML (1200 mg/kg),
having percentage chemosuppression of 54.5 ± 0.02, 76.9 ± 0.02, and
2.6. Test for curative activity (Rane’s test) 60.9 ± 0.01, respectively showed significantly lower suppressive ac­
tivities than DHP (2.18:17.45 mg/kg) and PA (4.58:1.53 mg/kg), with
Curative activities of APM and its components: MI, AI, NL and ML percentage chemosuppression of 88.1 ± 0.02 and 87.3 ± 0.02, respec­
were evaluated on established P. berghei infection in mice as described tively. However, the suppressive activity of AI (1240 mg/kg; percentage
by Ryley and Peters (1970). The mice were inoculated, grouped and chemosuppression of 92 ± 0.03) was comparable to those of DHP and
dosed as stated in section 2.4. The different treatments were started 72 h PA.
after the infection and were administered as a single dose per day for five
consecutive days. For the entire period of treatment (days 3–7), blood 3.2. Curative activity
samples were collected from tail snips of the mice daily and were made
into films. The blood films were developed for microscopic examination In established plasmodial infection in mice (72 h p. i.), all the herbal
to monitor their parasite densities as described above. decoctions: APM, NL, MI, ML, and AI; and positive controls: DHP and PA
showed chemosuppressive activities (Fig. 2 … (Fig. 2a and b). and
2.7. Test for prophylactic activity (Repository test) supplementary file 2). The different treatments significantly (p < 0.05)
reduced parasitaemia level from 24 h post administration to the end of
Prophylactic activities of APM and its components: MI, AI, NL and the treatment period (days 4–7) when compared to the negative control
ML were assessed in mice using the method described by Peters (1965). group. Throughout this period, the activities of all the herbal decoctions
The mice were inoculated, grouped and dosed as described in section were significantly lower than those of DHP and PA. At the termination of
2.4. The different treatments were administered as a single dose per day the treatment, DHP gave a total clearance (100%) of the parasites, PA,
for three consecutive days (days 0–2) before infection. On the fourth day AI, MI, ML, APM (320, 160, 100 mg/kg) and NL gave 99.5 ± 0.1%, 93.6
(day 3), the mice were inoculated and 72 h later (day 6), blood samples ± 0.04%, 93.3 ± 0.05%, 91.9 ± 0.02%, 91 ± 0.05%, 85.1 ± 0.04%, 82.5
were collected and the procedure completed as described above to ± 0.04%, and 72 ± 0.05% parasite clearance, respectively.
monitor their parasite densities.
3.3. Prophylactic activity
2.8. Data analysis
All the herbal decoctions: APM, NL, MI, ML, and AI exerted signifi­
All values are expressed as mean ± standard error of mean (SEM, cant (p < 0.05) reduction in parasitaemia level relative to the negative
with n = 5) and were analysed on GraphPad Prism Version 6.0 control in the prophylactic test (Fig. 3 and supplementary file 3). APM
(GraphPad Software, San Diego, USA). Statistical differences between suppressed parasitaemia level by 86.8 ± 0.03%, 79.1 ± 0.04%, and 60.8
the controls and treated groups were evaluated using one-way analysis ± 0.04% at doses of 320, 160, and 100 mg/kg, respectively; while ML,
of variances (ANOVA) with post hoc Tukey’s test; p values < 0.05 were MI, AI, and NL produced suppressive effect of 74.6 ± 0.03%, 71.1 ±
considered as being statistically significant. 0.02%, 67.1 ± 0%, and 35.8 ± 0.01%, respectively (see Fig. 3).

3. Results 4. Discussion

3.1. Suppressive activity Malaria remains one of the most prevalent infectious diseases in the
tropical regions of the world. More than a century after identification of
As shown in Fig. 1 (and supplementary file 1), APM; its components: the causative parasites and more than half a century after finding
NL, MI, ML, and AI; and positive controls: DHP and PA all showed effective drugs and insecticides, this dreadful disease is still yet to be
chemosuppressive activities on P. berghei parasitaemia in mice. These eradicated (Mehta and Desai, 2013). While there have been continuous
effects were statistically significant (p < 0.05) compared to the negative efforts for development of novel antimalarial drugs and effective vac­
control group. All doses of the polyherbal mixture (APM) significantly cines; at present there is no single drug effective for the treatment of

3
S.C. Alaribe et al. Journal of Ethnopharmacology 277 (2021) 114105

Fig. 2. (a). Curative effect of antimalarial decoctions and controls against established P. berghei infection in mice, showing parasite/μl. Each data point represents the
mean ± SEM; n = 5. Values in each group are significantly different (p < 0.05) from the other groups. (b). Curative effect of antimalarial decoctions and controls
against established P. berghei infection in mice, showing parasite clearance (%). Each data point represents the mean ± SEM; n = 5. Values in each group are
significantly different (p < 0.05) from the other groups.

multidrug resistant malaria; combination therapy has been the chemo­ models; suggesting that such preparations could be considered for
therapeutic strategy. Unfortunately, there has been report of develop­ further studies as an alternative strategy in our fight against malaria. In
ment of Plasmodium resistance to the WHO recommended this study we assessed and evaluated the suppressive, curative, and
artemisinin-based combination therapies (ACTs) in some parts of the prophylactic antiplasmodial activities of an antimalarial polyherbal
world (Dondorp et al., 2009). Some studies (Adepiti et al., 2014; Tar­ mixture (APM) in comparison to the activities of its individual constit­
kang et al., 2014; Orabueze et al., 2018) have shown that polyherbal uent plants and those of standard antimalarial drugs. The polyherbal and
preparations produced considerable antimalarial activities in animal individual plant decoctions were prepared in water and administered

4
S.C. Alaribe et al. Journal of Ethnopharmacology 277 (2021) 114105

suppression of parasitaemia is the most reported parameter. A per­


centage suppression of parasitaemia ≥10% relative to the vehicle con­
trol usually indicates that the test candidate is active (Peter and Anatoli,
1998). This suggests that all the decoctions investigated in this study
could be considered as active antimalarial candidates. Furthermore, in
vivo antiplasmodial activity is classified as moderate, good, or very good
if an extract showed percentage parasitaemia suppression ≥50% at a
dose of 500, 250 and 100 mg/kg/day, respectively (Munoz et al., 2000).
Based on this classification, the polyherbal mixture (APM) showed very
good antiplasmodial activities, NL showed good activities while MI, ML
and AI could be considered to have shown moderate activities. The
suppressive and curative activities demonstrated might be through in­
direct boosting of the immune system (Waako et al., 2005), as evidenced
by the daily decrease in parasitaemia level observed in the negative
control group in the curative model. The prophylactic activity might be
due to inhibition of proliferation of plasmodial parasites as a result of
direct cytotoxicity (Golenser et al., 2006) and modulation of erythrocyte
membranes to prevent parasite invasion (Hansen, 2012).
Fig. 3. Prophylactic activity of antimalarial decoctions and control against These activities could be attributed to the bioactive compounds
P. berghei infection in mice. Each bar represents the mean ± SEM; n = 5. Values present in the different decoctions. Studies have shown the presence of
in each group are significantly different (p < 0.05) from the other groups. alkaloids, terpenes, phenols, flavonoids, tannins, and saponins in NL,
MI, ML and AI (Okwu and Ezenagu, 2008; Bassey and Jude, 2014; Abu
orally to maintain the traditional methods of preparation and adminis­ et al., 2016; Nwali et al., 2018). These compounds could have elicited
tration. Four weeks old mice were chosen for the study in order to the observed antiplasmodial activities either individually or in syner­
exclude the effects of anaemia, which is likely in older mice (Pierrot gistic interactions or multi-factorial effects between themselves (Duke
et al., 2003). The in vivo models were adopted because they take into and Bogenschutz-Godwin, 1999; Gilbert and Alves, 2003); thus, isola­
consideration the possible prodrug effect and probable involvement of tion and investigation of the individual compounds is often necessary.
the immune system in controlling the infection (Waako et al., 2005). Many compounds have been isolated from stem bark of M. indica, the
The four day suppressive test usually assesses the antimalarial ac­ major component is the C-glucoside xanthone mangiferin (Fig. 4a). The
tivity of candidates on early periods of infection (Verma et al., 2011). In reported activities of mangiferin include immune-modulatory, antioxi­
this test, all the herbal decoctions significantly suppressed parasite level. dant and anti-inflammatory effects (Sanchez et al., 2000; Garcia et al.,
The polyherbal mixture (APM) gave the highest effect and showed 2002; Garrido et al., 2004). The immunostimulatory and
percentage chemosuppression in the range of 95.4–98.7% at the doses of anti-inflammatory actions of mangiferin could have contributed to the
100–320 mg/kg when administered 3 h after infection. Each of the in­ observed antimalarial effects of M. indica stem bark (Adepiti et al.,
dividual constituent plants (NL, MI, ML, and AI) also showed consider­ 2014). Compounds previously isolated from stem bark of A. indica
able suppressive effect (>50%), corroborating previous findings on include the diterpenoids margolone (Fig. 4d), margolonone (Fig. 4e),
these plants. Aqueous extract of N. latifolia roots was reported to exert and isomargolonone (Fig. 4b), which were reported to possess antibac­
85.22% suppressive effect at 100 mg/kg in early infection of P. berghei terial property (Ara et al., 1989). The limonoids nimbolide (Fig. 4f) and
(Alaribe et al., 2020); while aqueous fraction of the stem bark exhibited gedunin (Fig. 4h) are the most promising compounds from A. indica with
67.71% at 200 mg/kg (Ettebong et al., 2015). Similarly, MI, ML, and AI antimalarial property, and were isolated from the seed oil (Rojanapo
produced 78.2%, 67.72%, and 66.47% at 400 mg/kg, 400 mg/kg, and et al., 1985; Khalid et al., 1989). Compounds belonging to different
1000 mg/kg, respectively (Awe et al., 1998; Usha Devi et al., 2001; phytochemical classes have been isolated from the stem bark and root of
Umar et al., 2013). These indicate that the decoction of either the in­ N. latifolia. However, the alkaloids angustoline (Fig. 4c), naucleidinal
dividual plants or the combination could be useful in suppressing ma­ (Fig. 4g) and strictosamide (Fig. 4k) are the components reported to
laria in its early stages. . In the curative test, both DHP and PA showed exhibit antiplasmodial activity (Abreu and Pereira, 2001; Sun et al.,
remarkable activities. At the end of the treatment, DHP had totally 2008). Anthraquinones rubiadin 1-methyl ether (Fig. 4i) and dam­
cleared the parasites, with no measurable parasite recorded, while PA nacanthal (Fig. 4j) previously isolated from the root of M. lucida have
had elicited 99.5% parasite clearance. Typically, malaria parasite den­ been reported to exhibit potent antiplasmodial activity, producing
sities are expected to be negative 48 h after treatment with an ACT 98.7% and 100% inhibition of P. falciparum, respectively, at 30 μg/ml
(White, 2008). In a previous study, out of 63 patients diagnosed with (Koumaglo et al., 1992).
P. falciparum, 37 (58.7%) showed cleared parasitaemia at day 2 or 3 Wagner and Ulrich-Merzenich (2009) noted that individual com­
after treatment with DHP (Lo et al., 2016). All the herbal decoctions pounds rarely have the same degree of activity as the unrefined extract
were also active in the curative test. Significant reduction in parasite at comparable concentrations or doses. This phenomenon could be
load was observed from day 2 of treatment and daily for the five days of attributed to the heterogeneous nature of extracts in comparison to in­
treatment for each of the decoctions. At the end of treatment, none of the dividual compounds. Rasoanaivo et al. (2011) identified that crude ex­
decoctions had completely cleared the parasites, however they all pro­ tracts could benefit from positive pharmacodynamic and/or
duced substantial clearance (>70%), indicating their potential useful­ pharmacokinetic synergies between different constituents leading to
ness against established plasmodial infection. Similarly, in the activity at different receptor targets and/or improved pharmacokinetic
prophylactic test, the herbal decoctions demonstrated significant che­ profile of the main active principle(s). The authors further noted that
mosuppression. Although none of them completely suppressed para­ other beneficial interactions could include complementary mechanisms
sitaemia, they all (except for NL; 35%) produced notable of action (such as immunomodulation), reversal of resistance, and
chemosuppressive effect (>60%), implying that these decoctions could modulation of adverse effects. These phenomena could explain the
be applied as prophylactics for malaria. greater activities of APM relative to its individual constituent plants in
The suppressive, curative, and prophylactic antiplasmodial models both suppressive and prophylactic antiplasmodial models. Similar
are the standard methods commonly used for screening antimalarial studies have also shown that combining different medicinal plants or
candidates and in these methods, the determination of percentage orthodox drugs with herbal preparations could elicit beneficial effects

5
S.C. Alaribe et al. Journal of Ethnopharmacology 277 (2021) 114105

Fig. 4. Structures of some compounds isolated from M. indica (a), A. indica (b, d, e, f, h), N. latifolia (c, g, k) and M. lucida (i, j)

(Mohd Ridzuan et al., 2007; Anagu et al., 2014; Adepiti et al., 2014, org/10.1016/j.jep.2021.114105.
2016). Nevertheless, such polyherbal or herb-drug combinations could
also elicit antagonistic or subtractive effect or produce no significant Author statement
difference, as previously reported (Gathirwa et al., 2008; Idowu et al.,
2014). Although our findings indicate that the polyherbal mixture A:devised the project, the main conceptual ideas and supervised the
(APM) possessed antimalarial property and could be useful as treatment work
or prophylactic for malaria. Further studies are needed to evaluate the A&B:processed the experimental data, performed the analysis,
safety of this decoction and its activity in human. In the meantime, this drafted the manuscript and designed the figures.
study provides evidence supporting the notion that the use of stan­ A.C.D:worked out almost all of the technical details and carried out
dardized herbal medicines could be a valid complementary approach for the experiment.
malaria control, provided that the safety and efficacy of such formula­ A .B. & E: Discussed the results, aided in interpreting the results and
tions are clinically validated. worked on the manuscript.
F:Assisted with the technical staff on anti-plasmodial studies and
5. Conclusion data interpretation.

The results of this study revealed that the Antimalarial polyherbal References
mixture (APM) and its individual constituent plants (NL, MI, ML, and AI)
showed significant suppressive, curative and prophylactic antimalarial Abdulla, S., Salim, N., Machera, F., Kamata, R., Juma, O., Shomari, M., 2013.
Randomized, controlled trial of the long term safety, immunogenicity and efficacy of
activities. APM produced greater activities than its individual constitu­ RTS,S/AS02(D) malaria vaccine in infants living in a malaria-endemic region. Malar.
ent plants in both suppressive and prophylactic models, suggesting that J. 12 (11).
the combination produced synergistic or additive effects. This could Abreu, P., Pereira, A., 2001. New alkaloids from Sarcocephalus latifolius. Nat. Prod. Lett.
15, 43–48.
serve as scientific basis for the traditional usage of these plants as Abu, N.E., Ezeomeke, S.I., Azegba, P., Davidson, G.I., 2016. Phytochemical, nutritional
combined therapy for malaria. Further studies are recommended to and anti-nutritional properties of leaves, stems bark and roots of trees used in
evaluate their safety, disposition and interactions in human. popular medicine for the treatment of malaria in South Eastern Nigeria. J. Med.
Plants Res. 10 (38), 662–668.
Adebayo, J.O., Krettli, A.U., 2011. Potential antimalarials from Nigerian plants: a review.
Acknowledgement J. Ethnopharmacol. 133, 289–302.
Adejo, G.O., Atawodi, S.E., 2014. Acute toxicity and genotoxic effects of all parts of
Morinda lucida benth on pUC18 plasmid DNA. Nat. Prod. Chem. Res. S1, 006.
We appreciate the technical assistance of Mrs Chinonye Nwosu
Adepiti, A.O., Elujoba, A.A., Bolaji, O.O., 2014. In vivo antimalarial evaluation of MAMA
Anabike of the ANDI Centre of Excellence for Malaria Diagnosis, decoction on Plasmodium berghei in mice. Parasitol. Res. 113, 505–511.
Department of Medical Microbiology and Parasitology, College of Adepiti, A.O., Elujoba, A.A., Bolaji, O.O., 2016. Evaluation of herbal antimalarial MAMA
Medicine, University of Lagos. decoction-amodiaquine combination in murine malaria model. Pharm. Biol. 54 (10),
2298–2303.
Alaribe, C.S., Oladipupo, A.R., Musah, A.A., Coker, H.A.B., 2018. Investigation of
Appendix A. Supplementary data selected metallic impurities in antimalarial herbal decoctions ‘Agbo Iba’ collected
from four locations in Lagos state, Nigeria. UNILAG J. Med., Sci. Technol. 6 (1),
45–56.
Supplementary data to this article can be found online at https://doi.

6
S.C. Alaribe et al. Journal of Ethnopharmacology 277 (2021) 114105

Alaribe, C.S., Oladipupo, A.R., Nani, M., Ijeoma, I., Olanipekun, B., Coker, H.A.B., 2020. Mohd Ridzuan, M.A., Sow, A., NooRain, A., Mohd Ilham, A., Zakiah, I., 2007. Eurycoma
Suppressive and curative antiplasmodial properties of Nauclea latifolia root extract longifolia extract-artemisinin combination parasitaemia suppression of Plasmodium
and fractions against erythrocytic stage of mice-infective chloroquine-sensitive yoelii infected mice. Trop. Biomed. 24, 111–118.
Plasmodium berghei NK-65. J. Med. Plants Econ. Develop. 4 (1), 1–6 a72. Moreno, A., Joyner, C., 2015. Malaria vaccine clinical trials: what’s on the horizon. Curr.
Anagu, O.L., Attama, A.A., Okore, V.C., Gugu, H.T., Ngene, A.A., Esimone, C.O., 2014. Opin. Immunol. 35, 98–106.
Azadirachta indica extract-artesunic acid combination produces an increased cure Munoz, V., Sauvain, M., Bourdy, G., Callapa, J., Bergeron, S., Rojas, I., 2000. A search for
rate of Plasmodium berghei-infected mice. Pharm. Biol. 52, 883–889. natural bioactive compounds in Bolivia through a multidisciplinary approach. Part I.
Ara, I., Siddiqui, B.S., Faizi, S., Siddiqui, S., 1989. Structurally novel diterpenoid Evaluation of the antimalarial activity of plants used by the Chacobo Indians.
constituents from the stem bark of Azadirachta indica (melieceae). J. Chem. Soc. J. Ethnopharmacol. 69, 127–137.
Perkin. Trans. 2, 343–345. National Research Council NRC, 1996. Guide for the Care and Use of Laboratory
Awe, S.O., Olajide, O.A., Oladiran, O.O., Makinde, J.M., 1998. Antiplasmodial and Animals, eight ed. The National Academies Press, Washington DC, USA.
antipyretic screening of Mangifera indica extract. Phytother Res. 12, 437–438. Nwali, O.N., Idoko, A., Okolie, J.E., Ezeh, E., Ugwudike, P.O., Rita, O.N., Ezenwali, M.O.,
Bassey, S.A., Jude, E.O., 2014. Phytochemical composition and antidiabetic activity of Odo, I.A., Ani, P.N., Okolie, S.O., 2018. Comparative analysis of the phytochemical
ethanol root extract of Nauclea latifolia. J. Phytopharm. 3 (1), 52–56. compositions of leaf, stem-bark and root of Azadirachta Indica (neem). Univers. J.
Bejon, P., Lusingu, J., Olotu, A., Leach, A., Lievens, M., Vekemans, J., 2008. Efficacy of Pharm. Res. 3 (5), 46–50.
RTS,S/AS01E vaccine against malaria in children 5 to 17 months of age. N. Engl. J. Okpanyi, S.N., Ezeukwu, G.C., 1981. Anti-Inflammatory and antipyretic activities of
Med. 359 (24), 2521–2532. Azadirachta indica. J. Med. Plants Res. 41, 34–39.
Butler, D., Stricker, L., 2019. Promising Malaria Vaccine to Be Tested in First Large Field Okwu, D.E., Ezenagu, V., 2008. Evaluation of the phytochemical composition of mango
Trial. http://www.nature.com/articles/d41586-019-01232-4. (Accessed 13 (Mangifera Indica linn) stem bark and leaves. Int. J. Chem. Sci. 6 (2), 705–716.
September 2020). Orabueze, C.I., Adesegun, S.A., Ota, D.A., Coker, H.A., 2018. In vivo antiplasmodial
Chidozie, V.N., Adoga, G.I., Chukwu, O.C., Chukwu, I.D., Adekeye, A.M., 2014. activities of four Nigerian plants used singly and in polyherbal combination against
Antibacterial and toxicological effects of the aqueous extract of Mangifera Indica Plasmodium berghei infection. Indian J. Tradit. Knowl. 17 (4), 716–723.
stem bark on albino rats. Glob. J. Biol., Agricult. Health Sci. 3 (3), 237–245. Peter, I.T., Anatoli, V.K., 1998. The Current Global Malaria Situation. Malaria Parasite
Dondorp, A.M., Nosten, F., Yi, P., Das, D., Phyo, A.P., Tarning, J., Lwin, K.M., Arley, F., Biology, Pathogenesis, and Protection. ASM Press, Washington DC, USA, pp. 11–22.
Hanpithakpong, W., Lee, S.J., 2009. Artemisinin resistance in Plasmodium falciparum Peters, W., 1965. Drug resistance in Plasmodium berghei. Vincke and Lips, 1948; I.
malaria. N. Engl. J. Med. 361, 455–467. Chloroquine resistance. Exp. Parasitol. 17, 80–89.
Duke, J.A., Bogenschutz-Godwin, M.J., 1999. The synergy principle at work in plants, Pierrot, C., Adam, E., Lafitte, S., Godin, C., Dive, D., Capron, M., Khalife, J., 2003. Age-
pathogens, insects, herbivores and humans. In: Kaufmann, P.B., Cseke, L.J., related susceptibility and resistance to Plasmodium berghei in mice and rats. Exp.
Warbler, S., Duke, J.A., Brielmann, H.L. (Eds.), Natural Products from Plants. CRC Parasitol. 104, 81–85.
Press, Boca Raton, pp. 183–205. Rasoanaivo, P., Wright, C.W., Willcox, M.L., Gilbert, B., 2011. Whole plant extracts
Ettebong, E.O., Ubulom, P.M., Ekpenyong, C.E., Ekong, U.S., Akpan, O.E., Tambari, V., versus single compounds for the treatment of malaria: synergy and positive
2015. In vivo antiplasmodial activities of Nauclea latifolia. Asian J. Med. Sci. 6 (3), interactions. Malar. J. 10 (Suppl. 1), S4.
6–11. Rojanapo, W., Suwanno, S., Somaree, R., Glinsukon, T., Thebtaranonth, Y., 1985.
Fidock, D., Rosenthal, J., Croft, S., Brun, R., Nwaka, S., 2004. Antimalarial drug Screening of antioxidants from some Thia vegetables and herbs. J. Sci. Thailand 11,
discovery: efficacy models for drug screening. Nat. Rev. Drug Discov. 3, 509–520. 177–188.
Garcia, D., Delgado, R., Ubeira, F.M., Leiro, J., 2002. Modulation of rat macrophage Ryley, J.F., Peters, W., 1970. The antimalarial activity of some quinone esters. Ann. Trop.
function by the Mangifera indica L extracts Vimang and mangiferin. Int. Med. Parasitol. 84, 209–222.
Immunopharm. 2, 797–806. Sanchez, G.M., Re, L., Giuliani, A., Nunez-Selles, A.J., Davison, G.P., Leon-Fernandez, O.
Garrido, G., Delgado, R., Lemus, Y., Rodriquez, J., Garcia, D., Nunez-Selles, A.J., 2004. In S., 2000. Protective effects of Mangifera indica L extract, mangiferin and selected
vivo and in vitro anti-inflammatory activity of Mangifera indica L extract (VIMANG). antioxidants against TPA-induced biomolecules oxidation and peritoneal
Pharmacol. Res. 50, 165–172. macrophage activation in mice. Pharmacol. Res. 42, 565–573.
Gathirwa, J.W., Rukunga, G.M., Njagi, E.N., Omar, S.A., Mwitari, P.G., Guantai, A.N., Sun, J., Lou, J., Dai, S., Xu, H., Zhao, F., Liu, K., 2008. Indole alkaloids from Nauclea
Tolo, F.M., Kimani, C.W., Muthaura, C.N., Kirira, P.G., 2008. The in vitro officinalis with weak antimalarial activity. Phytochemistry 69, 1405–1410.
antiplasmodial and in vivo anti-malarial efficacy of combinations of some medicinal Taïwe, G.S., Ngo Bum, E., Talla, E., Dimo, T., Weiss, N., Sidiki, N., Dawe, A., Moto, F.C.
plants used traditionally for treatment of malaria by the Meru community in Kenya. O., Dzeufiet, P.D., De Waard, M., 2011. Antipyretic and antinociceptive effects of
J. Ethnopharmacol. 115, 223–231. Nauclea latifolia root decoction and possible mechanisms of action. Pharm. Biol. 49
Gilbert, B., Alves, L.F., 2003. Synergy in plant medicines. Curr. Med. Chem. 10, 13–20. (1), 15–25.
Golenser, J., Waknine, J.H., Krugliak, M., Hunt, N.H., Grau, G.E., 2006. Current Tarkang, P.A., Franzoi, K.D., Lee, S., Lee, E., Vivarelli, D., Freitas-Junior, L., Liuzzi, M.,
perspectives on the mechanism of action of artemisinins. Int. J. Parasitol. 36, Tsabang, N., Ayong, L.S., Agbor, G.A., Okalebo, F.A., Guantai, A.N., 2014. In vitro
1427–1441. antiplasmodial activities and synergistic combinations of differential solvent extracts
Greenwood, B.M., Armstrong, J.R.M., 1991. Comparison of two simple methods for of the polyherbal product. Nefang BioMed. Res. Int. 835013.
determining malaria parasite density. Trans. R Soc. Trop. 85 (2), 186–188. Umar, M.B., Ogbadoyi, E.O., Ilumi, J.Y., Salawu, O.A., Tijani, A.Y., Hassan, I.M., 2013.
Guinovart, C., Aponte, J.J., Sacarlal, J., Aide, P., Leach, A., Bassat, Q., 2009. Insights into Antiplasmodial efficacy of methanolic root and leaf extracts of Morinda lucida. J. Nat.
long-lasting protection induced by RTS,S/AS02 A malaria vaccine: further results Sci. Res. 3 (2), 112–122.
from a phase IIb trial in Mozambican children. PloS One 4, e5165. US Food and Drug Administration USFDA, 2005. Guidance for Industry: Estimating the
Hansen, D.S., 2012. Inflammatory responses associated with the induction of cerebral Maximum Safe Starting Dose in Adult Healthy Volunteers. US Food and Drug
malaria: lessons from experimental murine models. PLoS Pathog. 8, e1003045. Administration, Rockville, MD.
Idowu, O.A., Babalola, A.S., Adenubi, O.T., Olukunle, J.O., 2014. Antiplasmodial Usha Devi, C., Valecha, N., Atul, P.K., Pilla, i C.R., 2001. Antiplasmodial effect of three
activities of combined Extracts of Morinda morindiodes, Morinda lucida and Vernonia medicinal plants: a preliminary study. Curr. Sci. 80 (8), 917–919.
amygdalina in Plasmodium berghei infected mice. Zool. 11, 40–45. Verma, G., Dua, V.K., Agarwal, D.D., Atul, P.K., 2011. Anti-malarial activity of
Khalid, S.A., Duddeck, H., Gonzalez-Sierra, M., 1989. Isolation and characterization of Holarrhena antidysenterica and Viola canescens, plants traditionally used against
antimalerial agent of the neem tree, A. indica. J. Nat. Prod. 52, 922–927. malaria in the Garhwal region of north-west Himalaya. Malar. J. 10 (1), 20.
Knight, D.J., Peters, W., 1980. The antimalarial action of N-benzyloxy dihydrotriazines. Waako, P.J., Gumede, B., Smith, P., Folb, P.I., 2005. The in vitro and in vivo antimalarial
The action of cycloguanil (BRL50216) against rodent malaria and studies on its activity of Cardiospermum halicacabum and Momordica foetida. J. Ethnopharmacol.
mode of action. Ann. Trop. Med. Parasitol. 74, 393–404. 99 (1), 137–143.
Kouadio, J.H., Bleyere, M.N., Kone, M., Dano, S.D., 2014. Acute and sub-acute toxicity of Wagner, H., Ulrich-Merzenich, G., 2009. Synergy research: approaching a new
aqueous extract of Nauclea latifolia in Swiss mice and in OFA rats. Trop. J. generation of phytopharmaceuticals. Phytomedicine 16, 97–110.
Pharmaceut. Res. 13 (1), 109–115. Wan, Y.D., Zang, Y.D., Wang, J.S., 1992. Studies on the antimalarial action of gelatin
Koumaglo, K., Gbeassor, M., Nkabou, C.L., de Souza, C.C., Wenee, W., 1992. Effects of capsule of Artemisia annua. Zongguo ji sheng chong xue yu ji sheng chong bing za shi
three compounds extracted from Morinda lucida on Plasmodium falciparum. Planta (Chin. J. Parasitol. Parasitic Dis.) 10, 290–294.
Med. 58, 533–534. White, N.J., 2008. Qinghaosu (artemisinin): the price of success. Science 320, 330–334.
Lo, E., Nguyen, J., Oo, W., Hemming-Schroeder, E., Zhou, G., Yang, Z., Cui, L., Yan, G., World Health Organization WHO, 2015. Global Technical Strategy for Malaria 2016-
2016. Examining Plasmodium falciparum and P. vivax clearance subsequent to 2030. World Health Organization, Geneva.
antimalarial drug treatment in the Myanmar-China border area based on World Health Organization WHO, 2018. World Malaria Report, 2018. World Health
quantitative real-time polymerase chain reaction. BMC Infect. Dis. 16, 154. Organization, Geneva.
Mehta, D., Desai, N.J., 2013. Laboratory diagnosis of malaria-various method and its Wright, C.W., Linley, P.A., Brun, R., Wittlin, S., Hsu, E., 2010. Ancient Chinese methods
comparison. Natl. J. Integrated Res. Med. 4 (3), 138–143. are remarkably effective for the preparation of artemisinin-rich extracts of Qing Hao
with potent antimalarial activity. Molecules 15, 804–812.

You might also like