Open access peer-reviewed chapter

Stable Artesunate Resistance in A Humanized Mouse Model of Plasmodium falciparum

Written By

Sheetal Saini, Rajinder Kumar and Rajeev K. Tyagi

Submitted: 14 July 2021 Reviewed: 09 September 2021 Published: 12 October 2021

DOI: 10.5772/intechopen.100381

From the Edited Volume

Plasmodium Species and Drug Resistance

Edited by Rajeev K. Tyagi

Chapter metrics overview

275 Chapter Downloads

View Full Metrics

Abstract

Plasmodium falciparum, the most devastating human malaria parasite, confers higher morbidity and mortality. Although efforts have been made to develop an effective malaria vaccine, stage- and species-specific short-lived immunity crippled these efforts. Hence, antimalarial drug treatment becomes a mainstay for the treatment of malaria infection in the wake of the unavailability of an effective vaccine. Further, there has been a wide array of antimalarial drugs effective against various developmental stages of P. falciparum due to their different structures, modes of action, and pharmacodynamics as well as pharmacokinetics. The development of resistance against almost all frontline drugs by P. falciparum indicates the need for combination therapy (artemisinin-based combination therapy; ACT) to treat patients with P. falciparum. A higher pool of parasitemia under discontinuous in vivo artemisinin drug pressure in a developed humanized mouse allows the selection of artesunate resistant (ART-R) P. falciparum. Intravenously administered artesunate, using either single flash doses or a 2-day regimen, to the P. falciparum-infected human blood chimeric NOD/SCID.IL-2Rγ−/− immunocompromised (NSG) mice, with progressive dose increments upon parasite recovery, was the strategy deployed to select resistant parasites. Parasite susceptibility to artemisinins and other antimalarial compounds was characterized in vitro and in vivo. P. falciparum has shown to evolve extreme artemisinin resistance as well as co-resistance to antimalarial drugs. Overall, the present information shall be very useful in devising newer therapeutic strategies to treat human malaria infection.

Keywords

  • artemisinin
  • artesunate resistance
  • co-resistance
  • Plasmodium falciparum
  • humanized mouse model

1. Introduction

1.1 Malaria biology

Malaria is a leading parasitic disease caused by protozoans belonging to the genus Plasmodium (P.) when injected by the female mosquito (Anopheles) in humans during a blood meal. Out of 172 species of Plasmodium parasite, only five species are reportedly known to cause malaria infection: Plasmodium falciparum, Plasmodium malariae, Plasmodium vivex, Plasmodium ovale, and Plasmodium knowlesi. The parasite completes its sexual and asexual cycles in a very complicated manner [1]. Plasmodium gametocytes are taken up during the blood meal from the human host and travel through salivary glands to the gut of Anopheles and continue the sexual multiplication. The haploid male and female gametes fertilize and give rise to zygote in the gut, which further grows and elongates into the ookinetes. Ookinetes following several replication processes become oocyst that transforms into sporozoites [2]. These sporozoites move from the gut to salivary glands and are released by the mosquitoes into the human bloodstream during a blood meal.

Once sporozoites are released in the bloodstream, the asexual development stage gets started [3]. Sporozoites traverse to their first incubation site, that is, hepatocytes (pre-erythrocytic stage), and later continues the asexual blood-stage infection inside red blood cells (RBCs). The asexual blood-stage infection results in clinical manifestations of the disease. Some sporozoites reach the liver within a few hours and penetrate the hepatocyte and undergo asexual replication known as exo-erythrocytic schizogony. Each sporozoite develops into a schizont containing 18–26 merozoites. The entire pre-erythrocytic phase lasts about 5–16 days depending upon the parasite species. RBCs are “center stage” and host the asexual development of Plasmodium. The merozoites (infective form) released from the liver recognize, attach, and enter the RBCs quickly. Merozoite passes through a developmental transformation beginning with a characteristic ring stage and leading to the formation of a multinucleated schizont within RBCs. Mature schizont ruptures to release the merozoites and continues the multiplication in RBCs. Each mature schizont contains around 18–26 merozoites, and these are released following the lysis of RBC to invade fresh RBCs. It is the lysis of RBCs that induces the bouts of fever and anemia in the infected individual, whereas, in P. vivax and P. ovale infection, a few merozoites remain in dormant stages for four months or year causing malaria relapse [4].

1.2 Malaria vaccine

Globally, malaria is one of the leading causes of mortality (0.3–2.3%), and Africa and Asia are highly endemic regions among the other continents [5]. According to World Health Organization, approximately 299 million cases of malaria and 400,000 deaths have been reported worldwide in 2019 [6]. A variety of antimalarial drugs are available but the emergence of resistance has been a major setback in containing malaria infection. Currently, Mosquirix (RTS,S/AS01) developed by GlaxoSmithKline is the only vaccine available in the market against malaria. RTS,S/AS01 is an engineered vaccine comprising of the genes from the outer protein of P. falciparum circumsporozoites and a portion of a hepatitis B surface antigen with chemical adjuvant to boost the immune responses. Mosquirix reduced malaria cases in children to nearly half [7] and has had 40% efficacy in children receiving four vaccine doses [8]. However, the efficacy dropped to 26% in children receiving only three vaccine doses and 33% efficacy was observed during the first year in infants (up to 3 months old). Further, the effectiveness of Mosquirix is only for one year and failed to provide long-term protection but could be combined with chemotherapy to prevent the transmission of malaria in low-endemic regions [9].

A variety of vaccines focusing on irradiated sporozoites is under trial, which may give exposure to the array of antigens and help induce protective immunity against malaria [1].

1.3 Drug-based therapy

Antimalarial drugs are the most commonly used treatment option for malaria in tropical regions. Three types of antimalarial drugs are currently available viz. aryl amino alcohol compounds (quinine, lumefantrine, chloroquine, amodiaquine, mefloquine, etc.), antifolate compounds (pyrimethamine and sulfadoxine), and artemisinin and derivatives [10]. Plasmodium parasites have a complex life cycle that involves a mammalian and an invertebrate host. All the symptoms are caused by the repeated rupture and penetration of erythrocytes by the asexual blood-stage parasites (merozoites). Hence, most of the antimalarial drugs target the asexual erythrocytic stage of the parasite. As per WHO’s Model List of Essential Medicines (MLEM), currently, there are 14 curative drugs for the treatment of malaria (treatment postinfection) and 6 prophylactic medicines (treatment before infection), either single or in combination [11]. Curative drugs for P. falciparum are mostly artemisinin-based combinations with artemisinin derivative (short half-life) in combination with partner drug(s) with a different mechanism of action (longer half-life). Out of 14 curative drugs, chloroquine is used for P. vivax, primaquine is used for P. vivax and P. ovale both, and the rest 12 are used for treating P. falciparum malaria.

1.3.1 Quinine

First isolated from the bark of the cinchona tree in 1820, and it remained one of the most effective malaria treatment options till the early 2000s [12]. Even today, quinine is obtained entirely from its natural source due to its difficult synthesis of the active molecule. Resistance for quinine was first reported in the 1980s, and since 2006, not being used as a frontline antimalarial drug [12]. However, the drug is still present on the WHO’s list of essential medicines and is used wherever artemisinins are not available [11]. Quinine has blood schizonticidal and gametocytocidal activity against P. vivax and P. malariae. Quinine also inhibits heme polymerase activity (required to convert toxic heme into nontoxic hemazoin) and, hence, leads to the accumulation of heme (cytotoxic substrate) in parasites.

1.3.2 Chloroquine

It was used to treat all forms of malaria infections with fewer side effects in the 1940s [13]. Blood stage of P. vivax, P. ovale, and P. malariae, sensitive strains of P. falciparum, and gametocytes of P. vivax are sensitive to chloroquine. It is highly effective in controlling acute malaria infection as compared to quinine. Moreover, it has been efficient and safer to use to treat sensitive cases. The first resistance case was reported in the 1950s and by the time, many malaria parasites have developed resistance against chloroquine. As per MLEM, chloroquine is used as a curative and prophylactic drug against P. vivax in the regions where resistance is not known to evolve (Central American regions) [11].

1.3.3 Amodiaquine

It was first synthesized in 1948 [14] and used in combination with artesunate for treating uncomplicated P. falciparum malaria (Camoquine® or Coarsucam™) [15]. The mechanism of action of amodiaquine is similar to that of chloroquine involving inhibition of hemozoin formation [16].

1.3.4 Pyrimethamine and sulfadoxine

In the early 1950s, Elion G and Hitchings G developed pyrimethamine [17] and Elion, Hitchings, and Black won the joint Nobel Prize in Physiology or Medicine (1988) for “their discoveries of important principles for drug treatment.” Sulfadoxine was developed in the early 1960s [18] and in 1981, the pyrimethamine and sulfadoxine combination was approved for the treatment of malaria. The emergence of high levels of resistance against the combination of pyrimethamine and sulfadoxine led to its discontinuation as a prophylactic drug since both drugs inhibit the parasite’s folate biosynthesis pathway (dihydrofolate reductase, DHFR and dihydropteroate synthetase, respectively) [19].

1.3.5 Primaquine

Primaquine, an 8-aminoquinoline, was first used in early 1950s. 8-aminoquinolines eliminates mature P. falciparum gametocytes, exo-erythrocytic (hepatic) stage of all Plasmodium species, and prevents the relapse cases of P. vivax and P. ovale showing suboptimal blood-stage activity. Despite its remarkable antimalarial properties, primaquine is reportedly known to confer severe side effects [20].

1.3.6 Piperaquine

It was developed as a part of the Chinese National Malaria Elimination Programme in the 1960s [21]. Although China used this drug as a replacement for chloroquine, the emergence of resistance against piperaquine prohibited its use as monotherapy. Currently, this is used with a partner drug with DHA (Eurartesim®) as a combination therapy [22]. It binds to heme-containing species by blocking heme detoxification and acts through getting accumulated in the digestive vacuole [14].

1.3.7 Doxycycline

In the early 1960s, Pfizer Inc. (USA) invented doxycyclin, a synthetically derived broad-spectrum bacteriostatic agent from Streptomyces sp. Doxycyclin is an efficacious prophylactic drug and, in combination with a partner drug, is highly effective as a curative drug for Plasmodium infection. Doxycycline is particularly used as a preventive drug in the regions with chloroquine and multidrug-resistant (MDR) P. falciparum malaria. It is not recommended for pregnant women and children below 8 years of age, but adverse effects were scarcely reported [23].

1.3.8 Mefloquine

American Army developed mefloquine in the 1970s [24] and is still part of the MLEM. Initially introduced for the treatment of chloroquine-resistant malaria, mefloquine has been used as a curative (in combination with artesunate) and prophylactic drug. In the late 1980s, resistance cases were reported for mefloquine [25]. Further, antimalarial action is mediated by disrupting the hemoglobin digestion in the asexual erythrocytic stage of the parasite [14]. The rendered adverse effects on the central nervous system prohibited its use as an antimalarial drug [26].

1.3.9 Artemisinin and its derivatives

Tu Youyou first isolated artemisinin in 1971, from a traditional Chinese medicine plant Artemisia annua [27], and was conferred Nobel Prize (2015) in Physiology or Medicine for “her discoveries concerning a novel therapy against malaria.” Artemisinin and its derivatives (artemether, artesunate, and arteether) are metabolized to its active compound DHA and are effective against all MDR forms of P. falciparum. Artemisinins acts in multiple ways including the generation of free radicals after being activated by heme, which, in turn, destroys proteins essentially required for the parasite growth and development [28]. Additionally, its action is associated with upregulation of unfolded protein response (UPR) pathways [29] and downregulation of P. falciparum phosphatidylinositol-3-kinase (PfPI3K) [30] and Ca2+ transporter (PfATP6) [31]. Artemisinins are crucial to fight the battle against malaria with artemisinin combination therapy (ACT) accounting for the majority of current antimalarial treatments [22], and in the late 2000s, emerging artemisinin resistance was noticed in Southeast (SE) Asia [5, 32].

1.3.10 Lumefantrine

Chinese antimalarial research effort “Project 523” that led to the discovery of artemisinin also synthesized lumefantrine in 1976 [33]. Currently, lumefantrine is used in combination with artemether. Lumefantrine is known to inhibit the transcriptional and translational pathway of malaria parasites [16].

Advertisement

2. Artemisinin resistance

One of the greatest challenges in achieving malaria control is antimalarial drug resistance (Figure 1). It has been associated with the malaria dissemination to new areas and resurgence in areas where the disease had been eliminated from. The situation is worsened by the incomplete treatments as it puts massive drug selection pressure on P. falciparum parasites, and hence, it helps evolve resistance against all frontline drugs. Chloroquine resistance leads to the resurgence and spread of malaria for decades in most countries in the 1960s [34]. Pyrimethamine, amodiaquine (chloroquine analog), arylaminoalcohols mefloquine, and halofantrine too suffered reduced efficacy in the 1980s, spreading resistant parasites. WHO recommended the combined use of two or more compounds with different modes of action to provide necessary cure rates and delay the development and spread of resistance. With low mixed-strain transmission rates, Southeast (SE) Asia was historically the first region to show resistance to frontline drugs [32]. Resistance to chloroquine, mefloquine, and sulfadoxine-pyrimethamine was initially seen in SE Asia [35, 36]. Artemisinin-based combination therapies (ACTs) have been seen as effective in controlling malaria. ACTs were a first-line treatment option for malaria since the early 2000s and were quickly adopted worldwide [34].

Figure 1.

Timeline for the introduction of antimalarial drugs and emergence of resistance. Image illustrates the approximate timeline for the discovery of antimalarial drugs and evolution of resistance. In vitro and in vivo (huRBCs-reconstituted human blood chimeric/humanized mice) induction of artsunare resistance in laboratory strains of P. falciparum to confirm the emergence of resistance. CQ-R, chloroquine resistance; ART-R, artemisinin resistance; Q-R, quinine resistance; MF-R, mefloquine resistance.

The mechanism of action of most antimalarials depends on targeting a single pathway/molecule, for example, DHFR-mediated folate synthesis (sulfadoxine-pyrimethamine), cytochrome bc1 (atovaquone), and heme detoxification (chloroquine). The artemisinin(s) binds to an array of parasite proteins and influences multiple cellular processes including glycolysis, translational pathway, and cell cycle regulation [37, 38, 39, 40]. Some studies also suggest that artemisinin may target and depolarize the mitochondrial membrane potential [41, 42]. Due to these functional attributes, artemisinin eliminates the asexual blood stage and early sexual gametocyte forms of P. falciparum parasites at low concentrations (nM) regardless of its short half-life (<1 h). To overcome the half-life issue, artemisinin and its derivatives (artemether, artesunate, and dihydroartemisinin; DHA) are used in combination with a drug of longer half-life. In Southeast Asia (SE), primary artemisinin derivative combinations used are artesunate with mefloquine and DHA with piperaquine, whereas in Africa artemether with lumefantrine and artesunate and amodiaquine. In the late 2000s, emerging artemisinin resistance was noticed in SE Asia [5, 32] and the most problematic situation currently is the rapid increase in failure rates of DHA used in combination with piperaquine. Later have been the first-line treatment and the preferred ACT in most of the SE Asia [43, 44]. Artemisinin resistance enchants potential pressure on partner drugs to work quickly and effectively in case of efficacy failure of artemisinin. P. falciparum Kelch 13 (Pf-Kelch13) (located on chromosome 13) is shown to be primarily responsible for artemisinin resistance. Kelch13 protein is involved in intraerythrocytic parasite development and many other cellular processes including hemoglobin endocytosis responsible for parasite growth and antimalarial activity of artemisinin. K13 mutations in parasites mediate artemisinin resistance through the reduced killing potential of artemisinin drugs. Moreover, mutations in K13 drive the enhanced removal of damaged proteins by the parasite [45].

Triple artemisinin-based combination therapies (TACTs) and mass drug administration (MDA) are proposed to combat artemisinin resistance [46, 47]. Combining more than two drugs with different modes of action prevents the chances of multidrug resistance and its spread. MDA targets the asymptomatic malaria receptacles that may serve as hotbeds for the transmission and perseverance of resistant parasites. TACT efficacy is currently in the second phase; with underway Tracking Resistance to Artemisinin Collaboration II (TRAC II) multiple-site study. DHA, piperaquine and mefloquine, and artemether and lumefantrine along with amodiaquine combinations have shown promising results and might help to defer the evolution of artemisinin resistance. Moreover, TACTs might reinstate the artemisinin sensitivity in the areas prevalent for artemisinin resistance [48].

2.1 Experimental induction of artesunate resistance in vitro and in vivo (human RBCs-reconstituted NSG mice [humanized mice])

Various studies have been performed to understand artemisinin (ACT) resistance being evolved in laboratory strains of P. falciparum in vitro and in human blood chimeric mice. Human studies suggest single-dose artemisinin-induced dormant parasites in P. falciparum 3D7 or K13-infected strains of P. falciparum-infected patients. These parasites are most likely a reservoir for recrudescent infection following artemisinin mono- and combination therapy (ACT). Artemisinin-resistant P. falciparum may be experimentally selected following different regimens in vitro. The selected resistant parasites could employ different mechanism(s) of action to escape drug pressure for extended survival. Witkowski et al. [49] submitted laboratory P. falciparum (F32-Tanzania) to artemisinin pressure for over 3 years/100 cycles to select artemisinin-resistant parasites. These artemisinin-resistant parasite could survive up to 7000× of the initial IC50 of artemisinin (~10 nM) with unaltered chemosensitivity. Further, under high artemisinin pressure, parasites were arrested at the ring stage and re-gained their sensitive phenotype when drug pressure was removed. This unstable resistance phenotype questions the experimental generation of resistant phenotype [49]. Chavchich et al. [50] findings directly associated the development of resistance against artemisinin derivatives in P. falciparum strains (W2 and TM91C235) with the pfmdr1 gene and protein expression. However, there were no changes seen in these markers when D6 parasites were submitted to a similar drug pressure [50]. These findings were attested by Tucker et al. [51]. After continuous exposure of DHA for 1–2 months (320 nM maximum), Cui et al. were able to generate DHA-resistant P. falciparum strains 7G8, Dd2, HB3, and D10 but 3D7. DHA resistance was not seen limited to the ring stage but also occurred in trophozoites and schizonts like artemisinin [52]. Rocamora et al. [53] generated artemisinin-resistant P. falciparum parasite lines from 6A and 11C clones of the 3D7 strain of P. falciparum. Resistant clones displayed a significant decrease in artemisinin sensitivity within 1.5 months of selection and showed the enhanced cellular response against oxidative stress (antioxidant defense) and protein damage (unfolded protein response; UPR) [53]. Major pathways associated with UPR against artemisinin resistance are reported to be Plasmodium reactive oxidative stress complex (PROSC) and TCP-1 ring complex (TRiC) [29].

Rodrigues et al. [54] generated artesunate- and mefloquine-resistant P. chabaudi parasite in Balb/c mice and confirmed that resistance could be generated against combination drugs even when both drugs are administered simultaneously [54]. Maslachah et al. used P. berghei ANKA-infected Swiss mice and observed repeated passages of artemisinin-treated parasites in mice increased the effective dose of artemisinin from 50% to 90% with the reduced parasite clearance time and recrudescence time. Additionally, repeatedly artemisinin exposed parasites showed dormancy and vacuole formation [55]. Humans share >85% similarity with murines, but for the complexity of the cellular system and specificity of the human immune system, in vivo mouse models are far close to an ideal model for studying human malaria parasites. Rodent parasites are used as surrogates for human parasites, but the genetic differences between rodent and human parasites make it difficult to correlate with human studies. Human pathogens, which do not infect other animal species, require an animal model that could reconstitute or replicate the human immune system [56]. Fortunately, the mouse-human chimeric animals present a viable preclinical in vivo model to study the interaction of the human immune system with infectious agents. Immunodeficient mice engrafted with human RBCs (humanized mice) support the development of asexual blood-stage infection of P. falciparum [57, 58]. Artesunate-resistant (ART-R) P. falciparum (Uganada, Palo Alto Marburg) was experimentally selected by submitting discontinuous artesunate pressure on P. falciparum-infected humanized NOD/SCID IL-2Rγ−/− immunocompromised mice reconstituted with human erythrocytes (huRBC). This humanized animal model supported the incremental increase in artesunate dosage to select 100 times ART-R parasites (240 mg/kg) to that of clinical dose (2.4 mg/kg). ART-R phenotype exhibited two patterns of IC50 in the selected parasites in vivo. Further, the first-stage phenotype showed substantial resistance to artemisinin in vivo (400×) without a shift seen in IC50, and the second-stage resistance phenotype showed an absence of response to a very high artemisinin dose (3200×) with a shift in IC50 and co-resistance to quinine, halofantrine, and amodiaquine. This phenotype was further demonstrated as having high-grade and stable artemisinin resistance phenotype. This is the first report of its kind [59], wherein a humanized mouse model was developed and a stable ART-R phenotype was achieved. Moreover, mimicking the clinical double doe regime (for consecutive two days and 24 h apart), 41% survival was observed with the highest dose of artemisinin (80 mg/kg) in contrast to 80% survival in the single-dose protocol (240 mg/kg) (Figure 2) [59]. There have been humanized mouse models to study asexual blood and liver stage infection of P. falciparum [57, 58, 60, 61, 62] and P. vivax [63, 64].

Figure 2.

Experimental selection of artesunate-resistant P. falciparum in a huRBCs-reconstituted NSG mice (PfhuRBC-NSG). NOD.PrkdcscidIl2rg−/− (NSG) mice non-adaptively controlled residual immune responses and reconstituted with human RBCs (HuRBCs) were used to study of asexual blood-stage infection of P. falciparum. The artesunate resistance (ART-R) was developed by the stepwise selection in experimental humanized mice using single/flash dose (the highest dose: 240 mg/kg) and double-dose (the highest dose: 80 mg/kg) regimens. The stability of the resistance (ART-R) phenotype was attested in vivo by removing ART pressure for one month and then submitting the parasite to ART pressure. ART-R parasites exhibited co-resistance to quinine (QN), amodiaquine (AQ), and halofantrine (HF), and a combination of artesunate (AS) and AQ. These ART-R parasites showing cross-resistance were susceptible to chloroquine (CQ) and mefloquine (MQ).

Advertisement

3. Conclusion and future perspectives

Human malaria infection remains a biggest challenge to humanity and extensive research is en route to find newer therapeutic options to treat this systemic infectious disease, P. falciparum. Basic research has been focused to decipher the mechanisms of infection of Plasmodium species and biochemical, genomics, proteomics, and metabolomics pathways. This information is used to advance the applied research wherein incessant efforts are being made to design vaccines and drugs against this menace. An effective vaccine development against P. falciparum has been a cumbersome process as it takes longer durations than drug therapies. A variety of drugs are developed acting against all developmental stages of different species. The development of drug resistance is affected by various genetic factors (mutations in genes involved in drug transport and metabolism) as well as environmental factors (drug pressure, the geographical distribution of the parasite, etc.). Currently, artemisinin and its derivatives are the cornerstones of effective malaria therapy regimens. These drugs are used in “artemisinin-based combination therapy” for treating malarial infections where artemisinin or its derivatives are given in combination with another unrelated partner drug. Despite ACTs, a parasite is gaining resistance against almost all drugs at a frightening pace suggesting an urgent need to devise novel antimalarial drug(s). Advanced techniques in genomics and proteomics help in developing novel drugs and drug targets. Since single-drug therapy may lead to problems such as ineffective parasite clearance and development of resistance, newer drug combinations are also being developed to clear parasites at even lower doses.

In the end, we developed humanized mice (huRBCs-reconstituted NSG mice) and selected stable ART-resistant P. falciparum that showed co-resistance to amodiaquine, quinine, and halofantrine. If resistance to artesunate and artemisinins evolves at such a speed along with co-resistance to quinine and other antimalarials, we would be left with no satisfactory option for treating severe malaria and a compromised choice of treatments for uncomplicated malaria. Indeed, the current dependence on ARTs for both uncomplicated and severe malaria, together with a lack of viable therapeutic alternatives, is a compromising situation. We believe this may have dire consequences and would cripple efforts to achieve malaria control globally. Therefore, novel approaches are needed to devise newer drug and their targets to address this drug resistance issue.

Advertisement

Acknowledgments

Rajeev Tyagi would like to offer his sincere thanks to DBT, New Delhi, Govt. of India, for financially supporting this study in the form Ramalingaswami Re-entry Fellowship-2019 (D.O. NO. BT/HRD/35/02/2006) Sanction order (BT/RLF/Re-entry/27/2018).

Advertisement

Conflict of interests

Authors declare no conflict of interests exists. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Advertisement

Author’s contribution

RKT, SS, and RK contributed to conceptualization and writing; SS, RK, and RKT contributed to writing—review and editing. All authors have read and agreed to the final version of the manuscript.

References

  1. 1. Phillips M, Burrows J, Manyando C, et al. Malaria. Nature Reviews. Disease Primers. 2017;3:17050. DOI: 10.1038/nrdp.2017.50
  2. 2. Baker DA. Malaria gametocytogenesis. Molecular and Biochemical Parasitology. 2010;172:57-65
  3. 3. Cowman AF, Healer J, Marapana D, Marsh K. Malaria: Biology and disease. Cell. 2016;167:610-624
  4. 4. White NJ. Determinants of relapse periodicity in Plasmodium vivax malaria. Malaria Journal. 2011;10:297
  5. 5. White NJ, Pukrittayakamee S, Hien TT, et al. Malaria. Lancet. 2014;383:723-735
  6. 6. WHO/Malaria Key facts. 2021 Available from: https://www.who.int/news-room/fact-sheets/detail/malaria [Accessed: 07 July 2021]
  7. 7. RTS,S Clinical Trials Partnership (CTP). Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: Final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386:31-45
  8. 8. Penny MA, Verity R, Bever CA, et al. Public health impact and cost-effectiveness of the RTS,S/AS01 malaria vaccine: A systematic comparison of predictions from four mathematical models. Lancet. 2015;387:367-375
  9. 9. Gosling R, von Seidlein L. The future of the RTS,S/AS01 malaria vaccine: An alternative development plan. PLoS Medicine. 2016;13:e1001994
  10. 10. Golenser J, Waknine JH, Krugliak M, et al. Current perspectives on the mechanism of action of artemisinins. International Journal for Parasitology. 2006;36(14):1427-1441
  11. 11. WHO Model List of Essential Medicines. 2019. Available from: https://www.who.int/groups/expert-committee-on-selection-and-use-of-essential-medicines/essential-medicines-lists. [Accessed: 07 July 2021]
  12. 12. Achan J, Talisuna AO, Erhart A, et al. Quinine, an old anti-malarial drug in a modern world: Role in the treatment of malaria. Malaria Journal. 2011;10:144
  13. 13. Loeb F. Activity of a new antimalarial agent, chloroquine (SN 7618). Journal of the American Medical Association. 1946;130:1069-1070
  14. 14. Tse EG, Korsik M, Todd MH. The past, present and future of anti-malarial medicines. Malaria Journal. 2019;18:93
  15. 15. Bompart F, Kiechel JR, Sebbag R, Pecoul B. Innovative public-private partnerships to maximize the delivery of anti-malarial medicines: Lessons learned from the ASAQ Winthrop experience. Malaria Journal. 2011;10:143
  16. 16. Combrinck JM, Mabotha TE, Ncokazi KK, et al. Insights into the role of heme in the mechanism of action of antimalarials. ACS Chemical Biology. 2012;8:133-137
  17. 17. Russell PB, Hitchings GH. 2,4-Diaminopyrimidines as antimalarials. III. 5-Aryl derivatives. Journal of the American Chemical Society. 1951;73:3763-3770
  18. 18. Laing AB. Treatment of acute falciparum malaria with sulphorthodimethoxine (Fansil). British Medical Journal. 1965;1:905-907
  19. 19. Lumb V, Das MK, Singh N, et al. Multiple origins of Plasmodium falciparum dihydropteroate synthetase mutant alleles associated with sulfadoxine resistance in India. Antimicrobial Agents and Chemotherapy. 2011;55:2813-2817
  20. 20. Ashley EA, Recht J, White NJ. Primaquine: The risks and the benefits. Malaria Journal. 2014;13:418
  21. 21. Chen L, Qu FY, Zhou YC. Field observations on the antimalarial iperaquine. Chinese Medical Journal. 1982;95:281-286
  22. 22. Eastman RT, Fidock DA. Artemisinin-based combination therapies: A vital tool in efforts to eliminate malaria. Nature Reviews. Microbiology. 2009;7:864-874
  23. 23. Tan KR, Magill AJ, Parise M, Arguin PM. Centers for Disease Control and Prevention. Doxycycline for malaria chemoprophylaxis and treatment: Report from the CDC expert meeting on malaria chemoprophylaxis. The American Journal of Tropical Medicine and Hygiene. 2011;84(4):517-531
  24. 24. Trenholme C, Williams R, Desjardins R, et al. Mefloquine (WR 142,490) in the treatment of human malaria. Science. 1975;190:792-794
  25. 25. Brasseur P, Druilhe P, Kouamouo J, et al. High level of sensitivity to chloroquine of 72 Plasmodium falciparum isolates from Southern Cameroon in January 1985. The American Journal of Tropical Medicine and Hygiene. 1986;35:711-716
  26. 26. Nevin RL, Croft AM. Psychiatric effects of malaria and anti-malarial drugs: Historical and modern perspectives. Malaria Journal. 2016;15:332
  27. 27. Qinghaosu Antimalaria Coordinating Research Group (QACRG). Antimalarial studies on Qinghaosu. Chinese Medical Journal. 1979;92:811-816
  28. 28. Tilley L, Straimer J, Gnädig NF, et al. Artemisinin action and resistance in Plasmodium falciparum. Trends in Parasitology. 2016;32:682-696
  29. 29. Mok S, Ashley EA, Ferreira PE, et al. Drug resistance. Population transcriptomics of human malaria parasites reveals the mechanism of artemisinin resistance. Science. 2015;347(6220):431-435
  30. 30. Mbengue A, Bhattacharjee S, Pandharkar T, et al. A molecular mechanism of artemisinin resistance in Plasmodium falciparum malaria. Nature. 2015;520:683-687
  31. 31. Shandilya A, Chacko S, Jayaram B, Ghosh I. A plausible mechanism for the antimalarial activity of artemisinin: A computational approach. Scientific Reports. 2013;3:2513
  32. 32. Menard D, Dondorp A. Antimalarial drug resistance: A threat to malaria elimination. Cold Spring Harbor Perspectives in Medicine. 2017;7:a025619
  33. 33. Cui L, zhuan Su X. Discovery, mechanisms of action and combination therapy of artemisinin. Expert Review of Anti-Infective Therapy. 2009;7:999-1013
  34. 34. White NJ. Antimalarial drug resistance. Journal of Clinical Investigation. 2004;113:1084-1092
  35. 35. Roper C, Pearce R, Nair S, et al. Intercontinental spread of pyrimethamine-resistant malaria. Science. 2004;305:1124
  36. 36. Blasco B, Leroy D, Fidock DA. Antimalarial drug resistance: Linking Plasmodium falciparum parasite biology to the clinic. Nature Medicine. 2017;23:917-928
  37. 37. Shaw PJ, Chaotheing S, Kaewprommal P, et al. Plasmodiumparasites mount an arrest response to dihydroartemisinin, as revealed by whole transcriptome shotgun sequencing (RNA-seq) and microarray study. BMC Genomics. 2015;16:830
  38. 38. Wang J, Zhang CJ, Chia WN, et al. Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum. Nature Communications. 2015;6:10111
  39. 39. Ismail HM, Barton V, Phanchana M, et al. Artemisinin activity-based probes identify multiple molecular targets within the asexual stage of the malaria parasites Plasmodium falciparum 3D7. PNAS. 2016;113:2080-2085
  40. 40. Bridgford JL, Xie SC, Cobbold SA, et al. Artemisinin kills malaria parasites by damaging proteins and inhibiting the proteasome. Nature Communications. 2018;9:3801
  41. 41. Li W, Mo W, Shen D, et al. Yeast model uncovers dual roles of mitochondria in action of artemisinin. PLoS Genetics. 2005;1:e36
  42. 42. Wang J, Huang L, Li J, et al. Artemisinin directly targets malarial mitochondria through its specific mitochondrial activation. PLoS One. 2010;5:e9582
  43. 43. Phuc BQ, Rasmussen C, Duong TT, et al. Treatment failure of dihydroartemisinin/piperaquine for Plasmodium falciparum malaria, Vietnam. Emerging Infectious Diseases. 2017;23:715-717
  44. 44. van der Pluijm RW, Imwong M, Chau NH, et al. Determinants of dihydroartemisinin-piperaquine treatment failure in Plasmodium falciparum malaria in Cambodia, Thailand, and Vietnam: A prospective clinical, pharmacological, and genetic study. The Lancet Infectious Diseases. 2019;19:952-961
  45. 45. Kathryn JW, Mok S, Fidock DA. Molecular mechanisms of drug resistance in Plasmodium falciparum Malaria. Annual Review of Microbiology. 2020;74:431-454
  46. 46. Dini S, Zaloumis S, Cao P, et al. Investigating the efficacy of triple artemisinin-based combination therapies for treating Plasmodium falciparum malaria patients using mathematical modeling. Antimicrobial Agents and Chemotherapy. 2018;62:e01068-e01018
  47. 47. von Seidlein L, Peto TJ, Landier J, et al. The impact of targeted malaria elimination with mass drug administrations on falciparum malaria in Southeast Asia: A cluster randomised trial. PLoS Medicine. 2019;16:e1002745
  48. 48. van der Pluijm RW, Tripura R, Hoglund RM, et al. Triple artemisinin-based combination therapies versus artemisinin-based combination therapies for uncomplicated Plasmodium falciparum malaria: A multicentre, open-label, randomised clinical trial. Lancet. 2020;395:1345-1360
  49. 49. Witkowski B, Lelièvre J, Barragán MJ, et al. Increased tolerance to artemisinin in Plasmodium falciparum is mediated by a quiescence mechanism. Antimicrobial Agents and Chemotherapy. 2010;54(5):1872-1877
  50. 50. Chavchich M, Gerena L, Peters J, et al. Role of pfmdr1 amplification and expression in induction of resistance to artemisinin derivatives in Plasmodium falciparum. Antimicrobial Agents and Chemotherapy. 2010;54(6):2455-2464
  51. 51. Tucker MS, Mutka T, Sparks K, et al. Phenotypic and genotypic analysis of in vitro-selected artemisinin-resistant progeny of Plasmodium falciparum. Antimicrobial Agents and Chemotherapy. 2012;56(1):302-314. 21
  52. 52. Cui L, Wang Z, Miao J, et al. Mechanisms of in vitro resistance to dihydroartemisinin in Plasmodium falciparum. Molecular Microbiology. 2012;86(1):111-128
  53. 53. Rocamora F, Zhu L, Liong KY, et al. Oxidative stress and protein damage responses mediate artemisinin resistance in malaria parasites. PLoS Pathogens. 2018;14(3):e1006930
  54. 54. Rodrigues LA, Henriques G, Borges ST, et al. Experimental evolution of resistance to artemisinin combination therapy results in amplification of the mdr1 gene in a rodent malaria parasite. PLoS One. 2010;5(7):e11593
  55. 55. Maslachah L, Widiyatno TV, Yustinasari LR, Plumeriastuti H. Phenotypic approach artemisinin resistance in malaria rodent as in vivo model. Veterinary World. 2017;10(7):790-797
  56. 56. Rämer PC, Chijioke O, Meixlsperger S, et al. Mice with human immune system components as in vivo models for infections with human pathogens. Immunology and Cell Biology. 2011;89:408-416
  57. 57. Tyagi RK, Tandel N, Deshpande R, et al. Humanized mice are instrumental to the study of Plasmodium falciparum infection. Frontiers in Immunology. 2018;9:2550
  58. 58. Tyagi RK. Plasmodium falciparum infected humanized mice: A viable preclinical tool. Immunotherapy. 2021; DOI: 10.2217/imt-2021-0102
  59. 59. Tyagi RK, Gleeson PJ, Arnold L, et al. High-level artemisinin-resistance with quinine co-resistance emerges in P. falciparum malaria under in vivo artesunate pressure. BMC Medicine. 2018;16:181
  60. 60. Arnold L, Tyagi RK, Mejia P, et al. Analysis of innate defences against Plasmodium falciparum in immunodeficient mice. Malaria Journal. 2010;9:197
  61. 61. Arnold L, Tyagi RK, Meija P, et al. Further improvements of the P. falciparum humanized mouse model. PLoS ONE. 2011;6(3):e18045
  62. 62. Duffier Y, Lorthiois A, Cisteró P, et al. A humanized mouse model for sequestration of Plasmodium falciparum sexual stages and in vivo evaluation of gametocytidal drugs. Scientific Reports. 2016;6:35025
  63. 63. Minkah NK, Schafer C, Kappe SHI. Humanized mouse models for the study of human malaria parasite biology, pathogenesis, and immunity. Frontiers in Immunology. 2018;9:807
  64. 64. Schafer C, Roobsoong W, Kangwanrangsan N, et al. A humanized mouse Model for Plasmodium vivax to test interventions that block liver stage to blood stage transition and blood stage infection. IScience. 2020;23:101381

Written By

Sheetal Saini, Rajinder Kumar and Rajeev K. Tyagi

Submitted: 14 July 2021 Reviewed: 09 September 2021 Published: 12 October 2021