Submit manuscript...
eISSN: 2379-6367

Pharmacy & Pharmacology International Journal

Research Article Volume 10 Issue 6

Antimalarial evaluation of tamoxifen: A study in parasitized mice

Elias Adikwu,1 Simeon Ajeka Igono,2 Nwakaego Omonigho Ebong3

1Department of Pharmacology/Toxicology, Faculty of Pharmacy, Niger Delta University, Nigeria
2Department of Biology, Faculty of Natural and Applied Sciences, Ignatius Ajuru University of Education, Nigeria
3Department of Pharmacology/Toxicology, Faculty of Pharmacy, Madonna University, Nigeria

Correspondence: Elias Adikwu, Department of Pharmacology /Toxicology, Faculty of Pharmacy, Niger Delta University, Wilberforce Island, Bayelsa State, Nigeria, Tel 07068568868

Received: December 19, 2022 | Published: December 30, 2022

Citation: Adikwu E, Igono SA, Ebong NO. Antimalarial evaluation of tamoxifen: A study in parasitized mice. Pharm Pharmacol Int J. 2022;10(6):234-237. DOI: 10.15406/ppij.2022.10.00393

Download PDF

Abstract

Malaria parasites resistance to currently used antimalarial drugs is a clinical challenge, which adds to socio-economic burden. A quick and cost-effective solution is to discover new treatment alternatives through drug repurposing. Tamoxifen (TMX) is an anticancer drug with some discrepancies in documented antimalarial activity.  The current study assessed the in-vivo antiplasmodial activity of TMX on Plasmodium berghei-infected mice. Adult Swiss albino mice (both sexes) inoculated with Plasmodium berghei (1x107) intraperitoneally were used for curative and suppressive antiplasmodial studies. The inoculated mice were treated orally with TMX (1, 2 and 4 mg/kg/day) while the parasitized and standard controls were treated with normal saline (0.2mL/day) and chloroquine (CQ) (10mg/kg/day) for 4 days, respectively. Blood samples were collected and evaluated for parasitamia and haematological indices. The effects of TMX on body weight and rectal temperature were not significantly (p>0.05) different from the parasitized control. TMX did not produce significant (p>0.05) curative and suppressive antiplasmodial effects when compared to the parasitized control. Curatively, TMX at 1, 2 and 4 mg/kg produced 8.00 %, 14.39 % and 20.16 % parasitamia inhibitions, respectively compared to 79.21% parasitamia inhibition produced by CQ. In the suppressive study, 10.06 %, 17.44 % and 21.02 % parasitamia inhibitions were produced by TMX; 1, 2 and 4 mg/kg, respectively while CQ produced 82.10 % parasitamia inhibition. TMX had no significant (p>0.05) effects on red blood cells, white blood cells, packed cell volume and haemoglobin levels when compared to the parasitized control. This study showed that TMX lacks suppressive and curative antiplasmodial activities on Plasmodium berghei-infected mice.

Keywords: tamoxifen, repurpose, antimalaria, hematology, mice

Introduction

Malaria is a major public health threat in tropical and subtropical regions of the world. Despite the fact that less than 1% of malaria infections are fatal, it causes about 430,000 deaths per year, especially among young children in sub-Saharan Africa.1 Globally, 229 million cases of malaria and 409,000 deaths were reported in 2019.2 African countries account for about 94% cases of malaria and deaths globally while South-East Asia regions account for 3% cases of malaria infection2 One of the primary strategies for malaria management is early diagnosis and treatment. However, this strategy was progressively impaired by the emergence of resistance malaria parasites to antimalarial drugs including artemisinin based combination therapies (ACTs) the main stay for malaria therapy.3 Hence there is an urgent need for new and innovative treatments with novel targets to overcome the occurrence of resistance malaria parasites. One of the strategies to quickly and cost-effectively discover new treatment alternatives is to repurpose drugs approved for the treatment of other diseases.4 This approach has a very low risk of failure, because most clinically used drugs have been shown to be safe in humans. It is also less time consuming, therefore less investment is needed.4 Reliance on the traditional drug development pathways for new drugs has enormous implications on both cost and time.5

Tamoxifen (TMX) is a selective oestrogen-receptor modulator used for the treatment of oestrogen receptor positive breast cancer.6 Due to its low cost and safety profile, it is used worldwide.7 TMX has shown additional activities other than anticancer such as antibacterial and antiviral.6 It has also shown possible therapeutic benefits in parasitic infections, which include the inhibition of Taenia crassiceps activity in infected mice,8 Taenia solium cisticerci in hamsters9 and reduced Echinococcus granulosus survival.10 However, the effects of TMX on Plasmodium parasites remain conflicting. Studies showed it had no antiplasmodial effects on P. falciparum growth,11 Plasmodium yoelii nigeriensis-infected mice12 and chloroquine (CQ) resistant Plasmodium berghei-infected mice.13 But some studies documented in-vitro and in-vivo antiplaspmodial activities of TMX on Plasmodium falciparum and Plasmodium berghei (ANKA), respectively.14 Thus it is imperative that further studies are performed to ascertain the antiplasmodial activity of TMX.  The current study assessed the in-vivo antiplasmodial activity of TMX on Plasmodium berghei (NK65) - infected mice, which has no available literature.

Material and methods

Animals and drugs

TMX (Sigma Aldrich, St Louis, MO, USA) and CQ (Evans Pharm Nigeria Ltd) were used. The mice were purchased from the animal house of the Department of Pharmacology, Faculty of Basic Clinical Sciences, University of Port Harcourt, Rivers State, Nigeria.  The mice were housed in plastic cages under natural laboratory conditions. They were fed with food pellets and given water ad libitum. CQ sensitive Plasmodium berghei (P. berghei) (NK65) was sourced from Nigerian Institute of Medical Research, Yaba, Lagos. The mice were infected by intraperitoneal (i.p) inoculation of red blood cells containing P. berghei (1x107). Parasitamia was determined in Giemsa-stained thin blood smears by microscopy. TMX (1, 2 and 4 mg/kg).15 and CQ (10 mg/kg)16 were used. National Institute of Health Guidelines for the Care and Use of Laboratory Animals were followed.17

Curative antiplasmodial assessment of tamoxifen

The protocol on established malaria infection (4-day curative Test) was used.18 Thirty adult Swiss albino mice (both sexes) were inoculated as described above. The mice were grouped randomly into 6 of n=5/group and allowed for 3 days. On day 4, the mice were orally treated with TMX (1, 2 and 4 mg/kg/day) for 4 days, respectively.  The normal and parasitized controls were treated orally with normal saline (0.2mL/day) while the standard control was treated with CQ (10mg/kg/day) for 4 days, respectively.  Drops of tail blood were collected on slides daily and thin blood smears prepared. The blood smears were fixed with absolute methanol and stained with Giemsa stain (Sigma Aldrich, St Louis, MO, USA). Parasitamia levels were assessed using a light microscope. Percentage parasitamia and inhibitions were calculated as shown below.

Suppressive antiplasmodial assessment of tamoxifen

The protocol for early infection used was described by Peters.19 Thirty Swiss albino mice (both sexes) were inoculated as described above and randomly divided into 6 groups of 5 mice per group and allowed for 2hrs. Thereafter, the mice were orally treated with TMX (1, 2 and 4 mg/kg/day) for 4 days, respectively.  The normal and negative controls were treated orally with normal saline (0.2mL/day) while the standard control was treated with CQ (10mg/kg/day) for 4 days.  Tail blood samples were collected after treatment and thin blood smears were produced and processed as stated above. Percentage parasitamia and inhibitions were calculated as shown below.

% Parasitamia = Number of parasitized red blood cells ( RBCs )    Total number of RBCs count ×100% MathType@MTEF@5@5@+= feaagKart1ev2aaatCvAUfeBSjuyZL2yd9gzLbvyNv2CaerbuLwBLn hiov2DGi1BTfMBaeXatLxBI9gBaerbd9wDYLwzYbItLDharqqtubsr 4rNCHbGeaGqkY=Mj0xXdbba91rFfpec8Eeeu0xXdbba9frFj0=OqFf ea0dXdd9vqaq=JfrVkFHe9pgea0dXdar=Jb9hs0dXdbPYxe9vr0=vr 0=vqpWqaaeaabiGaciaacaqabeaadaqaaqaaaOqaaKqzGeaeaaaaaa aaa8qacaGGLaGaaiiOaiaadcfacaWGHbGaamOCaiaadggacaWGZbGa amyAaiaadshacaWGHbGaamyBaiaadMgacaWGHbGaaiiOaiabg2da9O WaaSaaaeaajugibiaad6eacaWG1bGaamyBaiaadkgacaWGLbGaamOC aiaabccacaWGVbGaamOzaiaabccacaWGWbGaamyyaiaadkhacaWGHb Gaam4CaiaadMgacaWG0bGaamyAaiaadQhacaWGLbGaamizaiaabcca caWGYbGaamyzaiaadsgacaqGGaGaamOyaiaadYgacaWGVbGaam4Bai aadsgacaqGGaGaam4yaiaadwgacaWGSbGaamiBaiaadohacaGGGcGc paWaaeWaaeaajugib8qacaWGsbGaamOqaiaadoeacaWGZbaak8aaca GLOaGaayzkaaqcLbsapeGaaiiOaiaacckacaGGGcaakeaajugibiaa dsfacaWGVbGaamiDaiaadggacaWGSbGaaeiiaiaad6gacaWG1bGaam yBaiaadkgacaWGLbGaamOCaiaabccacaWGVbGaamOzaiaabccacaWG sbGaamOqaiaadoeacaWGZbGaaeiiaiaadogacaWGVbGaamyDaiaad6 gacaWG0baaaiabgEna0kaaigdacaaIWaGaaGimaiaacwcaaaa@8EC9@

% Inhibition= ( % Parasitamia of negative control% Parasitamia of treated group )  % Parasitamia of negative control ×100% MathType@MTEF@5@5@+= feaagKart1ev2aaatCvAUfeBSjuyZL2yd9gzLbvyNv2CaerbuLwBLn hiov2DGi1BTfMBaeXatLxBI9gBaerbd9wDYLwzYbItLDharqqtubsr 4rNCHbGeaGqkY=Mj0xXdbba91rFfpec8Eeeu0xXdbba9frFj0=OqFf ea0dXdd9vqaq=JfrVkFHe9pgea0dXdar=Jb9hs0dXdbPYxe9vr0=vr 0=vqpWqaaeaabiGaciaacaqabeaadaqaaqaaaOqaaKqzGeaeaaaaaa aaa8qacaGGLaGaaeiiaiaadMeacaWGUbGaamiAaiaadMgacaWGIbGa amyAaiaadshacaWGPbGaam4Baiaad6gacqGH9aqpkmaalaaabaWdam aabmaabaqcLbsapeGaaiyjaiaacckacaWGqbGaamyyaiaadkhacaWG HbGaam4CaiaadMgacaWG0bGaamyyaiaad2gacaWGPbGaamyyaiaabc cacaWGVbGaamOzaiaabccacaWGUbGaamyzaiaadEgacaWGHbGaamiD aiaadMgacaWG2bGaamyzaiaabccacaWGJbGaam4Baiaad6gacaWG0b GaamOCaiaad+gacaWGSbGaeyOeI0IaaiyjaiaacckacaWGqbGaamyy aiaadkhacaWGHbGaam4CaiaadMgacaWG0bGaamyyaiaad2gacaWGPb GaamyyaiaabccacaWGVbGaamOzaiaabccacaWG0bGaamOCaiaadwga caWGHbGaamiDaiaadwgacaWGKbGaaeiiaiaadEgacaWGYbGaam4Bai aadwhacaWGWbaak8aacaGLOaGaayzkaaaapeqaaKqzGeGaaiiOaiaa cwcacaGGGcGaamiuaiaadggacaWGYbGaamyyaiaadohacaWGPbGaam iDaiaadggacaWGTbGaamyAaiaadggacaqGGaGaam4BaiaadAgacaqG GaGaamOBaiaadwgacaWGNbGaamyyaiaadshacaWGPbGaamODaiaadw gacaqGGaGaam4yaiaad+gacaWGUbGaamiDaiaadkhacaWGVbGaamiB aaaacqGHxdaTcaaIXaGaaGimaiaaicdacaGGLaaaaa@A40F@

Determination of body weight and rectal temperature

The mice in the curative study were weighed using a digital weighing balance. The pre- and post-treatment rectal temperature of the mice were measured using a digital rectal thermometer.

Evaluation of haematological parameters

Blood samples were collected from the mice used for the curative study and assessed for red blood cells (RBCs), white blood cells, (WBCs), packed cell volume (PCV) and hemoglobin (HB) concentrations using an auto analyser.

Determination of mean survival time

Mortality was monitored daily for each mouse in the control and experimental groups from the time of parasite inoculation up to death and was recorded in days. The mean survival time (MST) was calculated as expressed below. 

MST= Sum of survival time of all mice in group (days) Total number of mice in that group MathType@MTEF@5@5@+= feaagKart1ev2aaatCvAUfeBSjuyZL2yd9gzLbvyNv2CaerbuLwBLn hiov2DGi1BTfMBaeXatLxBI9gBaerbd9wDYLwzYbItLDharqqtubsr 4rNCHbGeaGqkY=Mj0xXdbba91rFfpec8Eeeu0xXdbba9frFj0=OqFf ea0dXdd9vqaq=JfrVkFHe9pgea0dXdar=Jb9hs0dXdbPYxe9vr0=vr 0=vqpWqaaeaabiGaciaacaqabeaadaqaaqaaaOqaaKqzGeaeaaaaaa aaa8qacaGGnbGaai4uaiaacsfacqGH9aqpkmaalaaabaGaam4uaiaa dwhacaWGTbGaaiiOaiaad+gacaWGMbGaaiiOaiaadohacaWG1bGaam OCaiaadAhacaWGPbGaamODaiaadggacaWGSbGaaiiOaiaadshacaWG PbGaamyBaiaadwgacaGGGcGaam4BaiaadAgacaGGGcGaamyyaiaadY gacaWGSbGaaiiOaiaad2gacaWGPbGaam4yaiaadwgacaGGGcGaamyA aiaad6gacaGGGcGaam4zaiaadkhacaWGVbGaamyDaiaadchacaGGGc WdaiaacIcapeGaamizaiaadggacaWG5bGaam4Ca8aacaGGPaaapeqa aiaadsfacaWGVbGaamiDaiaadggacaWGSbGaaiiOaiaad6gacaWG1b GaamyBaiaadkgacaWGLbGaamOCaiaacckacaWGVbGaamOzaiaaccka caWGTbGaamyAaiaadogacaWGLbGaaiiOaiaadMgacaWGUbGaaiiOai aadshacaWGObGaamyyaiaadshacaGGGcGaam4zaiaadkhacaWGVbGa amyDaiaadchaaaaaaa@8B20@

Statistical analysis

Graph pad prism version 5.02 was used to analyse data and expressed as mean ± standard error of mean. The differences between means were compared using one way analysis of variance (ANOVA) and Dunnet’s test. p<0.05 was considered significant.

Results

Effects of tamoxifen on body weight and rectal temperature of parasitized mice

In the curative study, treatment with TMX (1, 2 and 4 mg/kg) had no significant (p>0.05) effect on the body weight of parasitized mice when compared to the parasitized control. However, CQ significantly (p<0.05) restored the body weight of the parasitized mice when compared to the parasitized control (Table 1). TMX (1, 2 and 4 mg/kg) did not produce significant (p>0.05) effect on the rectal temperature of the parasitized mice when compared to the parasitized control. On the other, CQ significantly (p<0.05) increased rectal temperature of the parasitized mice when compared to the parasitized control (Table 2).

Treatment

BW (g) Day 1

BW (g) Day 4

BW Change

P C

25.01±4.21

22.77±3.44

-2.24a

CQ

24.33±3.60

25.34±2.32

+1.01b

TMX 1mg/kg

25.21±3.45

23.03±3.23

-2.18a

TMX 2 mg/kg

24.22±2.33

22.56±3.11

-2.16a

TMX 4 mg/kg

25.18±3.41

23.02±2.61

-2.10a

Table 1 Effect of tamoxifen on body weight of parasitized mice
Data as mean ± standard error of mean, n=5, BW, body weight; PC, parasitized control; CQ, chloroquine (Standard), TMX, tamoxifen. Values with difference superscripts down the column differ significantly at p<0.05 (ANOVA).

Treatment

TempºC

TempºC

TempºC

Day 1

Day 4

Difference

 P C

35.18±2.09

32.81±0.91

-2.62a

CQ

35.01±3.06

37.10±0.44

+2.09b

TMX 1 mg/kg

35.17±2.02

32.67±0.67

-2.50c

TMX 2 mg/kg

35.19±3.04

32.87±0.19

-2.32c

TMX 4mg/kg

35.04±2.08

32.90±0.55

-2.14c

Table 2 Effect of tamoxifen on temperature of parasitized mice
Data as mean ± standard error of mean, n=5, Temp, temperature; PC, parasitized control; CQ, chloroquine (Standard), TMX, tamoxifen. Values with difference superscripts down the column differ significantly at p<0.05 (ANOVA).

Curative antiplasmodial effect of tamoxifen on parasitized mice

TMX did not produce significant (p>0.05) curative antiplasmodial effect on day 1, 2, 3 and 4 of treatments when compared to the parasitized control (Tables 3 and 4). On the other hand, CQ showed significant (p<0.05) curative antiplasmodial activity on day 1, 2, 3 and 4 of treatments when compared to the parasitized control (Tables 3 and 4). On day 4 of treatment, TMX at 1, 2 and 4 mg/kg produced 8.00 %, 14.39 % and 20.16 % parasitamia inhibitions, respectively when compared to 79.21% parasitamia inhibition produced by CQ (Table 4). Treatment with TMX had no significant (p>0.05) effect on MST when compared to the parasitized control. On the other hand, CQ significantly (p<0.05) prolonged MST when compared to the parasitized control (Table 4).

Treatment

% Parasitamia Day 1

% Parasitamia Day 2

% Parasitamia Day 3

PC

25.74±2.23a

32.44±3.00b

42.75±2.62c

CQ

19. 43±2.00d

10.53±1.71e

5.12±0.56e

TMX 1mg/kg

22.24±2.47f

30.60±3.65g

39.61±3.42h

TMX 2mg/kg

22.64±2.73f

28.82±2.54g

37.65±3.00h

TMX 4mg/kg

21.39±2.54f

27.67±2.63g

35.33±2.81h

Table 3 Curative antiplasmodial effect of tamoxifen on daily parasitamia of parasitized mice
Data as mean ± standard error of mean, n=5, PC, parasitized control; CQ, chloroquine (Standard); TMX, tamoxifen. Values with difference superscripts differ significantly at p<0.05 (ANOVA: Analysis of variance).

Treatment

% Parasitamia

% Inhibition 

MST

PC

54.80±2.00a

0

9.30±0.46a

CQ

1.53±0.33b

79.21

30.56±2.33b

TMX 1mg/kg

50.42±3.52c

8.00

9.50±0.71c

TMX 2mg/kg

46.91±3.00c

14.39

9.63±0.23c

TMX 4 mg/kg

43.75±3.81c

20.16

9.80±0.34c

Table 4 Curative antiplasmodial effect tamoxifen on parasitized mice
Data as mean ± standard error of mean, n=5, PC, parasitized control; CQ, chloroquine (Standard); TMX, tamoxifen. Values with difference superscripts down the column differ significantly at p<0.05 (ANOVA: Analysis of variance).

Suppressive antiplasmodial effect of tamoxifen on parasitized mice

TMX (1, 2 and 4 mg/kg) had no significant (p>0.05) suppressive antiplasmodial effect when compared to the parasitized control (Table 5). However, CQ produced significant (p<0.05) suppressive antiplasmodial effect when compared to the parasitized control (Table 5). TMX at 1, 2 and 4 mg/kg produced 10.06 %, 17.44 % and 21.02 % parasitamia inhibitions, respectively while CQ produced 82.10 % parasitamia inhibition (Table 5). The effect of TMX on MST was not significantly (p>0.05) different from the parasitized control. However, MST was significantly (p<0.05) prolonged by CQ when compared to the parasitized control (Table 5).

Treatment

% Parasitamia

% Inhibition 

MST

PC

15.22±1.33a

0

9.60±0.66a

CQ

2.73±0.55b

82.10

35.56±2.11b

TMX 1mg/kg

13.60±1.33c

10.06

9.70±0.25c

TMX 2mg/kg

12.57±1.64c

17.44

10.00±0.54c

TMX 4 mg/kg

12.02±1.45c

21.02

10.63±0.11c

Table 5 Suppressive antiplasmodial effect of tamoxifen on parasitized mice
Data as mean ± standard error of mean, n=5, PC, parasitized control; CQ, chloroquine (Standard); TMX, tamoxifen. Values with difference superscripts down the column differ significantly at p<0.05 (ANOVA: Analysis of variance).

Effect of tamoxifen on haematological parameters of parasitized mice

P. berghei-infected mice showed (p<0.05) low levels of RBCs, Hb, PCV and high (p<0.05) levels of WBCs when compared to the normal control (Table 6). Treatment with TMX (1, 2 and 4 mg/kg) had no significant (p>0.05) effect on RBCs, Hb PCV and WBCs of parasitized mice when compared to the parasitized control (Table 6). On the other hand, CQ significantly (p<0.05) increased RBCs, Hb, PCV and significantly (p<0.05) decreased WBCs when compared to the parasitized control (Table 6).

Treatment

RBC (x106)

WBC (cells/L)

PCV (%)

HB (g/dL)

N C

6.07±0.54

3.86±0.63

59.34±4.45a

17.46±1.32a

 P C

3.21±1.32a

9.36±0.67a

31.33±3.00b

10.24±0.14b

CQ

5.21±0.72b

4.83±0.32b

54.33±5.42c

16.25±0.42c

TMX 1mg/kg

3.33±0.33c

8.45±0.45c

32.37±3.56c

10.33±0.71c

TMX 2mg/kg

3.46±0.25c

8.36±0.63c

33.47±3.43c

10.63±1.32c

TMX 4mg/kg

3.55±0.17c

8.24±0.22c

35.21±2.54c

11.00±0.52c

Table 6 Effect of tamoxifen on haematological parameters of parasitized mice
Data as mean ± standard error of mean, n=5, NC, normal control; PC, parasitized control; CQ, chloroquine (Standard); TMX, tamoxifen; RBCs, red blood cells; WBCs, white blood cells; PCV, packed cell volume; Hb, hemoglobin; Values with difference superscripts down the column differ significantly at p<0.05 (ANOVA).

Discussion

Regardless of the constant effort to reduce the burden of malaria infection, it was responsible for more than 260,000 deaths in children under 5 years in 2017.20 The emergence of new resistant strains of Plasmodium parasites to available antimalarial drugs21 is jeopardizing the international effort to combat malaria, which creates the need for the development of new therapies through drug repurposing. Drug repurposing is a very successful strategy, which accounts for around 25% of the annual income of the pharmaceutical industry.22 The selection of drugs for repurposing mostly considers clinically used drugs as well as drugs removed from the markets due to unprofitability or other strategic reasons.23 This study assessed the antimalarial activity of TMX on mice infected with CQ sensitive strain of P. berghei. A rodent model was used, because it produces disease features similar to those of human plasmodial infection when infected with P. berghei.24 CQ, which has been used in most antiplasmodial studies as a reference standard was used.25 P. berghei a rodent malaria parasite though, not able to infect man and other primates has been frequently used for the assessments of antimalarial drug candidates.25 In-vivo model was used in this study to allow for the possible prodrug effect and the eradication of infections by the immune system.26 Body weight loss and reduction in body temperature are symptoms of malaria-infected mice.27 In infected mice, decreased metabolic rate occurs before death and is accompanied by decreased internal body temperature.28,29 In this study, TMX did not produce notable effects on the body weight and rectal temperature of the parasitized mice. A 4-day curative study, which allows for established infection and a 4-day suppressive study, which allows for early infections were used.30 TMX exhibited no curative and suppressive antiplasmodial activities in this study. Similarly, Cervantes-Candelas et al.,13 reported that TMX had no antiplasmodial effect on CQ resistant P. berghei-infected mice. Staines et al.,11 also showed that TMX had no antiplasmodial effect on P. falciparum while Sing et al.,12 reported no antiplasmodial effect on P. yoelii nigeriensis-infected mice. In contrast to findings in the current study, Weinstock et al.,14 showed that TMX and its primary metabolite 4-HO-tamoxifen produced in-vitro and in- vivo antiplaspmodial activities on P. falciparum and P. berghei (ANKA) respectively. The observations in this study differ from findings by Weinstock et al.,14 probably due to differences in the dose of TMX, species of P. berghei and mice used. MST experimentally measures the ability of a potential antimalarial drug to prevent or reduced malaria associated death.30 Treatment with TMX did not prolong MST of P. berghei-infected mice.  Plasmodium parasites are common cause of anaemia especially in children leading to decreased RBCs, Hb and PCV. Malaria parasites use host Hb as an essential nutrient for multiplication and growth. Malaria parasites ingest more than 75% of Hb during their intra-erythrocytic phase and metabolize heme into hemozoin.31 One of the characteristic of an antimalarial drug candidate is to surmount anaemic condition caused by Plasmodium parasites. This study observed the inability of TMX to prevent P. berghei-induced anemia. In contrast, Cervantes-Candelas et al.,13 reported deceased anemia in P. berghei ANKA-infected mice treated with TMX.  

Conclusion

Tamoxifen did not produce antiplasmodial activity on P. berghei-infected mice.

Acknowledgments

None.

Conflicts of interest

Authors declare that there is no conflict of interest.

References

  1. Moxon CA, Gibbins MP, McGuinness D et al. New insight into malaria pathogenesis. Annu Rev Pathol. 2020;15:315‒343.
  2. World malaria report 2020. Geneva: WHO; 2020.
  3. Shibeshi MA, Kifle ZD, Atnafie SA. Antimalarial Drug Resistance and Novel Targets for Antimalarial Drug Discovery. Infect Drug Resist. 2020;13:4047‒4060.
  4. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: Progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18:41–58.
  5. Matthews H, Usman-Idris M, Khan F, et al. Drug repositioning as a route to anti-malarial drug discovery: preliminary investigation of the in vitro anti-malarial efficacy of emetine dihydrochloride hydrate. Malaria J. 2013;12:359.
  6. Bogush TA, Polezhaev BB, Mamichev IA et al. Tamoxifen never ceases to amaze: new fndings on nonestrogen receptor molecular targets and mediated effects. Cancer Invest. 2018;36(4):211–220.
  7. World Health Organization. Model list of essential medicines. 21st List; 2019.
  8. Vargas Villavicencio JA, Larralde C, Nava MADL, et al. Tamoxifen treatment induces protection in murine cysticercosis. J Parasitol. 2007;93(6):1512‒1517.
  9. Escobedo G, Palacios-Arreola MI, Olivos A, et al. Tamoxifen treatment in hamsters induces protection during taeniosis by Taenia soliumBiomed Res Int. 2013;2013:280496.
  10. Nicolao MC, Elissondo MC, Denegri GM, et al. In vitro and in vivo effects of tamoxifen against larval stage Echinococcus granulosusAntimicrob Agents Chemother. 2014;58(9):5146–5154.
  11. Staines HM, Dee BC, Shen MR et al. The effect of mefoquine and volume-regulated anion channel inhibitors on induced transport in Plasmodium falciparum-infected human red blood cells. Blood Cells Mol Dis. 2004;32(3):344–348.
  12. Singh N, Puri SK. Interaction between chloroquine and diverse pharmacological agents in chloroquine resistant plasmodium yoelii nigeriensis. Acta Trop. 2000;77(2):185–193.
  13. Cervantes-Candelas LA, Aguilar-Castro J, Orlando F, et al. Tamoxifen Suppresses the Immune Response to Plasmodium berghei ANKA and Exacerbates Symptomatology. Pathogens. 2021;10(6):743.
  14. Weinstock A, Gallego-Delgado J, Gomes C, et al. Tamoxifen activity against Plasmodium in vitro and in mice. Malar J. 2019;18(1):378.
  15. Oliveira RN, Correa SAP, Vieira KM, et al. In vitro schistosomicidal activity of tamoxifen and its effectiveness in a murine model of schistosomiasis at a single dose. Parasitol Res. 2019;118(5):1625–1631.
  16. Adikwu E, Ajeka IS. Artemether/lumefantrine/clindamycin eradicates blood and liver stages of Plasmodium berghei infection in mice. J Anal Pharm Res. 2021;10(6):240‒244.
  17. Guide for the Care and Use of Laboratory Animals. The National Academies Press: Washington, DC, USA, 8th edition; 2011.
  18. Ryley JF, Peters W. The antimalarial activity of some quinolone esters. Ann Trop Med Parasitol.1970;64(2):209‒222.
  19. Peters W. Rational methods in the search for antimalarial drugs. Transaction of Royal Society of Tropical Medicine and Hygiene. 1967;61(3):400‒410.
  20. World malaria report 2018. Geneva: World Health Organization; 2018.
  21. Hayton K, Ranford-Cartwright LC, Walliker D. Sulfadoxine-Pyrimethamine Resistance in the Rodent Malaria Parasite Plasmodium chabaud.Antimicrob Agents and Chem. 2002;46(8):2482‒2489.
  22. Talevi A, Bellera CL. Challenges and opportunities with drug repurposing: Finding strategies to find alternative uses of therapeutics. Expert Opin Drug Discov. 2020;15(4):397–401.
  23. Jourdan JP, Bureau R, Rochais C, et al. Drug repositioning: A brief overview. J Pharm Pharmacol. 2020;72(9):1145–1151.
  24. Alli LA, Adesokan AA, Salawu OA, et al. Anti-plasmodial activity of aqueous root extract of Acacia nilotica. African J Biochem Res. 2011;5(7):214-219.
  25. Adikwu E, Ajeka SI, Ebong N. In-vivo Antiplasmodial Impact of Dihydroartemisinin-Piperaquine- Clindamycin on Plasmodium berghei-Infected Mice. Am J Biomed Sci. 2022;14(1):29-38.
  26. Nardos A, Makonnen E. In vivo antiplasmodial activity and toxicological assessment of hydroethanolic crude extract of Ajuga remota. Malar J. 2017;16(1):25.
  27. Fentahun S, Makonnen E, Awas T, et al. In vivo antimalarial activity of crude extracts and solvent fractions of leaves of Strychnos mitis in Plasmodium berghei infected mice. BMC Complement Altern Med. 2017;17(1):13.
  28. Dikasso D, Makonnen E, Debella A et al. Anti-malarial activity of Withania somnifera L. dunal extracts in mice. Ethiop Med J. 2006;44(3):279–285.
  29. Saxena M, Saxena J, Nema R. Phytochemistry of medicinal plants. Journal of Pharmacognosy and Phytochemistry. 2013;1(6):168–182.
  30. Adikwu E, Ajeka IS, Nworgu CO. Amodiaquine-Azithromycin Eradicates Blood and Liver Stages of Plasmodium berghei Infection in Mice. Am J Biomed Sci. 2022;14(3):136‒145.
  31. Inbaneson SJ, Sundaram R, Suganthi P. In vitro antiplasmodial effect of ethanolic extracts of traditional medicinal plant Ocimum species against Plasmodium falciparumAsian Pac J Trop Med. 2012;5(2):103–106.
Creative Commons Attribution License

©2022 Adikwu, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.