28
Expanding the Antimalarial Drug Arsenal—Now, But How? Brian T. Grimberg and Rajeev K. Mehlotra Center for Global Health and Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Tel.: +1-216-368-6328 or +1-216-368-6172, Fax: +1-216-368-4825 Brian T. Grimberg: [email protected]; Rajeev K. Mehlotra: [email protected] Abstract The number of available and effective antimalarial drugs is quickly dwindling. This is mainly because a number of drug resistance-associated mutations in malaria parasite genes, such as crt, mdr1, dhfr/dhps, and others, have led to widespread resistance to all known classes of antimalarial compounds. Unfortunately, malaria parasites have started to exhibit some level of resistance in Southeast Asia even to the most recently introduced class of drugs, artemisinins. While there is much need, the antimalarial drug development pipeline remains woefully thin, with little chemical diversity, and there is currently no alternative to the precious artemisinins. It is difficult to predict where the next generation of antimalarial drugs will come from; however, there are six major approaches: (i) re-optimizing the use of existing antimalarials by either replacement/rotation or combination approach; (ii) repurposing drugs that are currently used to treat other infections or diseases; (iii) chemically modifying existing antimalarial compounds; (iv) exploring natural sources; (v) large-scale screening of diverse chemical libraries; and (vi) through parasite genome- based (“targeted”) discoveries. When any newly discovered effective antimalarial treatment is used by the populus, we must maintain constant vigilance for both parasite-specific and human- related factors that are likely to hamper its success. This article is neither comprehensive nor conclusive. Our purpose is to provide an overview of antimalarial drug resistance, associated parasite genetic factors (1. Introduction; 2. Emergence of artemisinin resistance in P. falciparum), and the antimalarial drug development pipeline (3. Overview of the global pipeline of antimalarial drugs), and highlight some examples of the aforementioned approaches to future antimalarial treatment. These approaches can be categorized into “short term” (4. Feasible options for now) and “long term” (5. Next generation of antimalarial treatment—Approaches and candidates). However, these two categories are interrelated, and the approaches in both should be implemented in parallel with focus on developing a successful, long-lasting antimalarial chemotherapy. Keywords malaria; falciparum; artemisinin resistance; natural products; drug discovery; kinases; HDAC; DHODH © 2011 by the authors; licensee MDPI, Basel, Switzerland. Correspondence to: Brian T. Grimberg, [email protected]; Rajeev K. Mehlotra, [email protected]. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/3.0/). NIH Public Access Author Manuscript Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26. Published in final edited form as: Pharmaceuticals (Basel). 2011 May 1; 4(5): 681–712. doi:10.3390/ph4050681. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

Expanding the Antimalarial Drug Arsenal—Now, But How?

Brian T. Grimberg and Rajeev K. MehlotraCenter for Global Health and Diseases, School of Medicine, Case Western Reserve University,Cleveland, OH 44106, USA; Tel.: +1-216-368-6328 or +1-216-368-6172, Fax: +1-216-368-4825Brian T. Grimberg: [email protected]; Rajeev K. Mehlotra: [email protected]

AbstractThe number of available and effective antimalarial drugs is quickly dwindling. This is mainlybecause a number of drug resistance-associated mutations in malaria parasite genes, such as crt,mdr1, dhfr/dhps, and others, have led to widespread resistance to all known classes of antimalarialcompounds. Unfortunately, malaria parasites have started to exhibit some level of resistance inSoutheast Asia even to the most recently introduced class of drugs, artemisinins. While there ismuch need, the antimalarial drug development pipeline remains woefully thin, with little chemicaldiversity, and there is currently no alternative to the precious artemisinins. It is difficult to predictwhere the next generation of antimalarial drugs will come from; however, there are six majorapproaches: (i) re-optimizing the use of existing antimalarials by either replacement/rotation orcombination approach; (ii) repurposing drugs that are currently used to treat other infections ordiseases; (iii) chemically modifying existing antimalarial compounds; (iv) exploring naturalsources; (v) large-scale screening of diverse chemical libraries; and (vi) through parasite genome-based (“targeted”) discoveries. When any newly discovered effective antimalarial treatment isused by the populus, we must maintain constant vigilance for both parasite-specific and human-related factors that are likely to hamper its success. This article is neither comprehensive norconclusive. Our purpose is to provide an overview of antimalarial drug resistance, associatedparasite genetic factors (1. Introduction; 2. Emergence of artemisinin resistance in P. falciparum),and the antimalarial drug development pipeline (3. Overview of the global pipeline of antimalarialdrugs), and highlight some examples of the aforementioned approaches to future antimalarialtreatment. These approaches can be categorized into “short term” (4. Feasible options for now)and “long term” (5. Next generation of antimalarial treatment—Approaches and candidates).However, these two categories are interrelated, and the approaches in both should be implementedin parallel with focus on developing a successful, long-lasting antimalarial chemotherapy.

Keywordsmalaria; falciparum; artemisinin resistance; natural products; drug discovery; kinases; HDAC;DHODH

© 2011 by the authors; licensee MDPI, Basel, Switzerland.Correspondence to: Brian T. Grimberg, [email protected]; Rajeev K. Mehlotra, [email protected] article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license(http://creativecommons.org/licenses/by/3.0/).

NIH Public AccessAuthor ManuscriptPharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

Published in final edited form as:Pharmaceuticals (Basel). 2011 May 1; 4(5): 681–712. doi:10.3390/ph4050681.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 2: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

1. Introduction1.1. Malaria

More than 40% of the world’s population, much of it socioeconomically and politicallychallenged, live in areas where malaria, alone or together with HIV/AIDS and tuberculosis,is a significant health risk [1,2]. According to the World Health Organization (WHO),approximately 250 million clinical cases of malaria occur every year [3]. Malaria isestimated to kill nearly one million people annually, with most of the deaths occurring inchildren under 5 years of age in sub-Saharan Africa [4]. If children survive multipleinfections, such exposure leads to a natural immunity that limits the severity of the diseaselater in life. However, this immunity wanes in the absence of continued exposure to malariainfections. Additionally, pregnant women and newborns have reduced immunity, andtherefore are vulnerable to severe complications of malaria infection and disease [5,6].

Malaria is primarily caused by four species of the protozoan parasite Plasmodium: P.falciparum, P. vivax, P. malariae, and P. ovale, which are transmitted by over 70 species ofAnopheles mosquitoes [7,8]. These parasite species occur sympatrically both in humanpopulations and within infected individuals, with P. falciparum and P. vivax being thepredominant species [9–11]. Approximately 80% of all malaria cases and 90% of malaria-attributed deaths occur in Africa, and are caused by P. falciparum [3]. Outside of Africa, P.vivax is the most widespread species, occurring largely in Asia, including the Middle Eastand the Western Pacific, and in Central and South America [12]. This parasite speciescauses a relatively less lethal form of the disease compared with P. falciparum [12,13].

1.2. Overview of Antimalarial DrugsToday when many mosquito vectors are resistant to insecticides [14,15], and an effectivevaccine is not yet available [14,16], chemoprophylaxis/chemotherapy remains the principalmeans of combating malaria. Over the past 60 to 70 years, since the introduction of syntheticantimalarials, only a small number of compounds, belonging to three broad classes, havebeen found suitable for clinical usage [17,18]. These classes are described below.

1.2.1. Quinine and related drugs—Quinine, originally extracted from cinchona bark inthe early 1800s, along with its dextroisomer quinidine, is still one of the most importantdrugs for the treatment of uncomplicated malaria, and often the drug of last resort for thetreatment of severe malaria. Chloroquine (CQ), a 4-aminoquinoline derivative of quinine,has been the most successful, inexpensive, and therefore the most widely used antimalarialdrug since the 1940s. However, its usefulness has rapidly declined in those parts of theworld where CQ-resistant strains of P. falciparum and P. vivax have emerged and are nowwidespread. Amodiaquine, an analogue of CQ, is a pro-drug that relies on its activemetabolite monodesethylamodiaquine, and is still effective in areas of Africa, but not inregions of South America. Other quinine-related, commonly used drugs include mefloquine,a 4-quinoline-methanol derivative of quinine, and the 8-aminoquinoline derivative,primaquine; the latter is specifically used for eliminating relapse causing, latent hepaticforms (hypnozoites) of P. vivax. Halofantrine and lumefantrine, both structurally related toquinine, were found to be effective against multidrug-resistant falciparum malaria[17,19,20]. While lumefantrine, in combination with an artemisinin derivative, artemether(Coartem), is recommended by the WHO for treating uncomplicated falciparum malaria,halofantrine generally is not recommended because of serious side effects and extensivecross-resistance with mefloquine [21].

1.2.2. Antifolate combination drugs—Antifolate drugs include various combinationsof dihydrofolate reductase enzyme (DHFR) inhibitors, such as pyrimethamine, proguanil,

Grimberg and Mehlotra Page 2

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 3: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

and chlorproguanil, and dihydropteroate synthase enzyme (DHPS) inhibitors, such assulfadoxine and dapsone. With rapidly growing sulfadoxine-pyrimethamine (SP) resistance,a new combination drug, Lapdap (chlorproguanil-dapsone), was tested in Africa in the early2000s, but was withdrawn in 2008 because of hemolytic anemia in patients with glucose-6-phosphate dehydrogenase enzyme (G6PD) deficiency [22].

1.2.3. Artemisinin and its derivatives—Artemisinin drugs, which originated from theChinese herb qing hao (Artemisia annua), belong to a unique class of compounds, thesesquiterpene lactone endoperoxides [18]. The parent compound of this class is artemisinin(quinghaosu), whereas dihydroartemisinin (DHA), artesunate, artemether, and β-arteetherare the most common derivatives of artemisinin; DHA is the main bioactive metabolite of allartemisinin derivatives (artesunate, artemether, β-arteether, etc.), and is also available as adrug itself [17,18,20].

Since 2001, the WHO has recommended the use of artemisinin-based combination therapies(ACTs) for treating falciparum malaria in all countries where resistance to monotherapies ornon-artemisinin combination therapies (e.g., SP) is prevalent [23]. The rationale for the useof ACTs is based on the facts that artemisinin derivatives are highly potent and fast acting,and that the partner drug in ACT has a long half-life, which allows killing the parasites thatmay have escaped the artemisinin inhibition. Thus, it is thought that ACTs will delay theonset of resistance by acting as a “double-edged sword” [18,24,25]. ACTs, such asartemether-lumefantrine, artesunate-amodiaquine, and artesunate-mefloquine are being usedin China, Southeast Asia, many parts of Africa, and some parts of South America[17,18,20]. Introduction of ACTs has initiated noticeable reduction in malaria prevalence inthese endemic regions of the world [18]. Although the mechanism(s) of action is poorlyunderstood (described below), the current high level of interest in artemisinin drugs is due totheir well-recognized pharmacological advantages: These drugs act rapidly upon asexualblood stages of CQ-sensitive as well as CQ-resistant strains of both P. falciparum and P.vivax, and reduce the parasite biomass very quickly, by about 4-logs for each asexual cycle.

In addition, these drugs are gametocytocidal. Thus, through rapid killing of asexual bloodstages and developing gametocytes, artemisinin drugs significantly limit the transmissionpotential of the treated infections. These drugs have large therapeutic windows, and basedon extensive human use, they appear to be safe, even in children and mid/late pregnantwomen. Furthermore, there is no reported “added toxicity” when these drugs are used incombination with other types of antimalarial compounds. In addition to these three mainclasses of compounds, the antibiotic tetracycline and its derivatives, such as doxycycline, areconsistently active against all species of malaria, and in combination with quinine, areparticularly useful for the treatment of severe falciparum malaria [17]. Until recently, acombination of atovaquone, a hydroxynaphthoquinone, and proguanil (Malarone) wasconsidered to be effective against CQ- and multidrug-resistant falciparum malaria;atovaquone resistance has recently been noticed in Africa [26]. Piperaquine, anothermember of the 4-aminoquinoline group, in combination with DHA (Artekin), holds thepromise of being successful in CQ-resistant endemic areas of Southeast Asia [17,18,20].

1.3. Overview of Antimalarial Drug ResistanceThis limited antimalarial armament is now severely compromised because of the parasite’sremarkable ability to develop resistance to these compounds [19,27]. In many differentmalaria-endemic areas, low to high-level resistance in the predominant malaria parasites, P.falciparum and P. vivax, has been observed for CQ, amodiaquine, mefloquine, primaquine,and SP. Plasmodium falciparum has developed resistance to nearly all antimalarial drugs incurrent use, although the geographic distribution of resistance to any one particular drug

Grimberg and Mehlotra Page 3

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 4: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

varies greatly. In particular, Southeast Asia has a highly variable distribution of falciparumdrug resistance; some areas have a high prevalence of complete resistance to multiple drugs,while elsewhere there is a spectrum of sensitivity to various drugs [19]. Until 2009, nonoticeable clinical resistance to artemisinin drugs was reported. However, as describedbelow, a number of recent studies have raised concerns about the efficacy of ACTs,particularly in Southeast Asia.

1.4. Overview of Genetic Basis for Antimalarial Drug ResistanceIt is believed that the selection of parasites harboring polymorphisms, particularly pointmutations, associated with reduced drug sensitivity, is the primary basis for drug resistancein malaria parasites [28,29]. Drug-resistant parasites are more likely to be selected if parasitepopulations are exposed to sub-therapeutic drug concentrations through (a) unregulated druguse; (b) the use of inadequate drug regimens; and/or (c) the use of long half-life drugs singlyor in non-artemisinin combination therapies. In recent years, significant progress has beenmade to understand the genetic/molecular mechanisms underlying drug resistance in malariaparasites [30,31]. Chloroquine resistance (CQR) in P. falciparum is now linked to pointmutations in the chloroquine resistance transporter (PfCRT [encoded by pfcrt, located onchromosome 7]). Pfcrt-K76T mutation confers resistance in vitro, and is the most reliablemolecular marker for CQR. Polymorphisms, including copy number variation and pointmutations, in another parasite transporter, multidrug resistance (PfMDR1 or Pgh1 [encodedby pfmdr1, located on chromosome 5]), contribute to the parasite’s susceptibility to a varietyof antimalarial drugs. Point mutations in pfmdr1 play a modulatory role in CQR, whichappears to be a parasite strain-dependent phenomenon [32].

Point mutations in the P. falciparum DHPS enzyme (encoded by pf-dhps, located onchromosome 8) are involved in the mechanism of resistance to the sulfa class ofantimalarials, and accumulation of mutations in the P. falciparum DHFR domain (encodedby pf-dhfr, located on chromosome 4) defines the major mechanism of high-levelpyrimethamine resistance. In field studies, a pf-dhps double mutant (437G with either 540Eor 581G), combined with the pf-dhfr triple mutant (108N_51I_59R), was found to befrequently associated with SP treatment failure [28,29]. Orthologues of pfcrt (pvcrt-o),pfmdr1 (pvmdr1), pf-dhps (pv-dhps), and pf-dhfr (pv-dhfr) in P. vivax have been identified,and found to be polymorphic. However, associations of the mutant alleles of pvcrt-o/pvmdr1and pv-dhps/pv-dhfr with clinical resistance to CQ and SP, respectively, are unclear [33].

2. Emergence of Artemisinin Resistance in P. falciparumA major advance in the search for effective treatment for drug-resistant malaria came withthe discovery of artemisinin and its derivatives. These compounds show very rapid parasiteclearance times and faster fever resolution than any other currently licensed antimalarialdrug [18]. Given the recent significant impact of ACTs on malaria morbidity and mortality,it is worrisome that higher recrudescence rates of P. falciparum malaria after artemisinintreatment are seen in some areas. Recrudescence, the reappearance of an infection after aperiod of quiescence, occurs in up to 30% of patients on artemisinin monotherapy, and in upto 10% of patients on ACTs [18,34]. The underlying mechanism of recrudescence afterartemisinin treatment is unclear. As illustrated by recent in vitro studies, the occurrence ofparasite dormancy, where parasites enter a temporary growth-arrested state, may provide aplausible explanation for this phenomenon [35,36].

Furthermore, there are recent concerns that the efficacy of ACTs has declined near the Thai-Cambodia border, a historical “hot spot” for emergence and evolution of multidrug-resistantmalaria parasites [37]. In this region, artemisinin resistance is characterized by significantlyslower parasite clearance in vivo, with or without corresponding reductions on conventional

Grimberg and Mehlotra Page 4

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 5: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

in vitro susceptibility testing [38,39]. A recent heritability study found that the observedartemisinin-resistant phenotype of the parasites has a genetic basis [40]. The study showedthat genetically identical parasite strains, defined by microsatellite typing, strongly clusteredin patients with slow versus faster parasite clearance rates. It was suggested that theartemisinin-resistant phenotype is expected to spread within parasite populations that livewhere artemisinins are deployed unless associated fitness costs of the putative resistancemutation(s) outweigh selective benefits. The genetic basis for artemisinin resistance is notknown at present. The main reason for this lack of knowledge is that the molecularmechanism(s) of action of artemisinin drugs remains uncertain and debatable, although anumber of potential targets have been proposed [41,42]. Investigations into potential proteintargets of artemisinin drugs have included PfATP6, a P. falciparum SERCA-type calcium-dependent ATPase in the endoplasmic reticulum [43]; PfCRT; PfMDR1; P. falciparummultidrug resistance-associated protein 1 (PfMRP1), residing on the parasite plasmamembrane; a putative deubiquitinating protease termed UBP-1; and mitochondrial proteins[41,42]. A recent study, conducted on artemisinin-resistant P. falciparum strains fromwestern Cambodia, did not find any correlation between artemisinin-resistant phenotype andpfmdr1 mutations/amplification, as well as mutations in pfatp6 (or pfserca), pfcrt, ubp-1, ormtDNA [44].

At present there is no concrete evidence of artemisinin resistance occurring elsewhere.Preliminary reports of emerging artemisinin resistance in South America and some Africancountries are as yet unconfirmed. For now, the situation appears to be confined to the Thai-Cambodia border, and is most likely a result of distinctive features of malaria treatment inthis geographic area. These include unregulated artemisinin monotherapy for more than 30years, creating massive drug pressure, and counterfeited or substandard tablets that containless active ingredients than stated [45]. However, if the problem is not urgently andappropriately addressed, it is feared that artemisinin resistance could spread worldwide.Given the pivotal role that artemisinin drugs now play, this would be a major blow formalaria treatment and control, since there are currently no alternative drugs to replace them.Even the development pipeline of new drugs consists predominantly of ACTs.

3. Overview of the Global Pipeline of Antimalarial DrugsOlliaro and Wells [46] reviewed the global pipeline of new antimalarial combinations andchemical entities, in various stages of development till February 2009. Overall, the pipelineconsists of 22 projects, either completed (artemether-lumefantrine and artesunate-amodiaquine), in various clinical stages (phase III/registration stage, n = 4; phase II, n = 8;phase I, n = 5), or in preclinical translational stage (n = 3). Chemical novelty is relativelylow among the products in clinical stages of development; the prevailing families are ofartemisinin-type and quinoline compounds, mostly in combination with each other. Amongthe non-artemisinin and/or non-quinoline combinations and single compounds are:Azithromycin-CQ (phase III), fosmidomycin-clindamycin (phase II), methylene blue-amodiaquine (phase II), SAR 97276 (a choline uptake antagonist, phase II), and tinidazole(an anti-parasitic nitroimidazole compound, phase II).

It is clear from the size of the antimalarial drug development pipeline that the pipeline hasnot reached critical mass, which is of concern particularly when we consider the recentemergence of artemisinin resistance, and the apparent decrease in time to resistance to eachnew drug/drug combination. The challenges for now and for the future are: (i) how to ensurethat we have compounds to combat emerging and future antimalarial drug resistance; and(ii) how to initiate and expedite the development of compounds that are as innovative aspossible with respect to their chemical scaffold and molecular target.

Grimberg and Mehlotra Page 5

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 6: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

4. Feasible Options for Now4.1. Re-Optimizing Existing Antimalarials

4.1.1. Replacing/rotating drugs—Examples of CQ and SP—Recently, a drugreplacement/rotation approach has been suggested to stem the tide of resistance by changingthe “playing field” for malaria parasites [47,48]. This approach is based on theunderstanding that the drug-resistant mutant forms are likely to be less fit than their wild-type strains in the absence of drug selection [49]. It has been shown that when CQ waswithdrawn from Malawi, where a high level of CQR prevailed, CQ sensitivity returned [50].The return of CQ-sensitive falciparum malaria in Malawi took almost 10 years, and wasassociated with a re-expansion of pfcrt-76K allele-carrying diverse, sensitive parasites[51,52]. Similar association between cessation of CQ use and decrease in pfcrt-76T allele-carrying resistant parasites has been observed in Kenya [53] as well as in China [54,55].However, it is estimated that in these areas, sensitivity to CQ may take many more years toreturn to a level where the drug could be used effectively again [53,54].

Regarding SP, it has been shown that in the Amazon region of Peru, the frequencies of themutant pf-dhfr and pf-dhps alleles, conferring SP resistance, declined significantly 5 yearsafter the drug was replaced [48]. This, however, was not the case in Venezuela [56] andCambodia [57], where the SP resistance-conferring alleles have remained at a highfrequency 8 years and 2 decades, respectively, after the replacement of SP. It appears thatthe outcome of the drug replacement/rotation approach is not universal but malaria-endemicregion dependent; a number of diverse ecological and epidemiological factors are likely toplay a role in determining the outcome of this approach. In addition, the logistics, includingthe cost component, of this approach may pose challenges for some countries.

4.1.2. Combining drugs—Examples of CQ-CQR reversers and multiple-drugtherapy—Improving the use of existing antimalarial drugs by designing new combinationsis another potential approach to combat resistance. CQ entered widespread usage in the1940s [19]. In spite of widespread resistance, this drug is still widely available because it isinexpensive and relatively non-toxic. In a thought-provoking article, Ginsburg [58]suggested that differential use of CQ in holoendemic areas that precludes children andpregnant women could still confer an efficacious protection for semi-immune adults, andmore efficient treatment protocols could be devised to treat even patients infected with CQ-resistant parasite strains. According to the author [58], since the antimalarial activity of CQis pleiotropic, drug resistance may be due to different mechanisms, each amenable toreversal by drug combination. Since the pivotal study by Martin and colleagues [59], whichshowed that CQR in P. falciparum was partly reversible by verapamil, it has beenenvisioned that the usefulness of CQ can be rescued by attempting to reverse the resistance—By co-administering CQ with another drug that chemosensitizes the parasite to its effect[60–64]. Among more than 40 such compounds, only one, chlorpheniramine, a histamineH1 receptor antagonist, in combination with CQ has been tested in patients with somesuccess [58,60]. Of further interest, primaquine and antiretroviral protease inhibitors (PIs)saquinavir, ritonavir, and indinavir, may act as CQR reversers [60]. Fluoxetine, anantidepressant of the selective serotonin reuptake inhibitor class, has been shown to reverseboth CQ and mefloquine resistance, while ketoconazole, a synthetic antifungal drug of theimidazole class, has been shown to reverse mefloquine resistance in a monkey model [60].In addition, novel compounds that are more potent and less toxic than the archetypal CQRreverser verapamil are under investigations [60]. Thus, the reports of new resistancereversers over the last few years have generated considerable interest, and may lead to aneffective stopgap therapy for drug-resistant malaria.

Grimberg and Mehlotra Page 6

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 7: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

Use of combination therapy is currently mandated by the WHO [23]. With widespread high-level CQR, SP, as a 2-drug combination, was a successful first-line treatment in Africa foralmost two decades. Currently, our hope rests mainly upon ACTs and atovaquone-proguanil(Malarone). As mentioned before, it is of concern that some of these combinations havestarted showing signs of failure. A further approach to minimize/delay the development ofdrug resistance in malaria is to apply combinations of three or more drugs (“multiple-drugtherapy”), as is practiced for treating HIV/AIDS and tuberculosis. Attempts at combiningexisting drugs with CQ (e.g., CQ-SP) have proved less successful, whereas amodiaquine-SPhas shown encouraging clinical activity [65]. Recently, a triple combination of artesunate-Lapdap, despite the side effects associated with Lapdap, is under evaluation for thetreatment of uncomplicated falciparum malaria in Africa [66]. A limitation of this approachis the increased possibility of drug-drug interactions or the need for a reformulation of thedrugs, both of which can prove costly and time-consuming to resolve. Furthermore, giventhat only a handful of effective antimalarial drugs are now left, this approach seems lesspromising.

4.1.3. (Re) Considering drugs with limitations—Example of tafenoquine—Withany drug, one of the major limitations is side effects. Mefloquine was first synthesized in1969 primarily for the purpose of chemoprophylaxis in the U.S. military following the thenrecently discovered threat of CQ-resistant falciparum malaria. The drug has been in use asmonotherapy and as combination therapy with either SP or artesunate. However, due toincreasing incidences of resistance, particularly in the areas of multidrug resistance [67,68],newer information about toxicity [69,70], and the potential for severe neuropsychiatricadverse events among US military personnel [71], doxycycline is considered as areplacement for mefloquine chemoprophylaxis [72]. Although doxycycline has scanty sideeffects, a major limitation is that in prophylactic therapy, it must be taken daily leading tohigh non-compliance. Therefore, a new investigational drug, tafenoquine, an analogue ofprimaquine, related to mefloquine, was introduced in 1990s [73,74]. Although likeprimaquine, tafenoquine causes hemolysis in patients with G6PD deficiency, the drug hasthe potential advantage of a shorter treatment course, and therefore increased patientcompliance, and higher therapeutic index than primaquine [73–75]. Thus, the interest intafenoquine has been continued and efforts are directed at ameliorating side effects, so thatthis new efficacious compound can be used for malaria prevention as well as treatment [76–78].

4.2. Repurposing Drugs Used for Other Infections or DiseasesOne of the approaches for rapidly identifying and bringing new drugs to the market for thetreatment of malaria is to repurpose existing drugs that are currently approved for treatmentof other infections or diseases.

4.2.1. Drugs used for other infections—Anti-HIV/AIDS drugs—It is known thatsome antimalarial drugs have moderate anti-HIV activities [79,80]. However, recent studieshave also demonstrated that certain antiretroviral agents can inhibit malaria parasite growth.Skinner-Adams et al. [81] showed that antiretroviral PIs saquinavir, ritonavir, and indinavirwere effective against P. falciparum. Subsequently, other antiretroviral PIs were also shownto exhibit potent antimalarial activity [82,83]. The current working hypothesis for theantimalarial activity of antiretroviral PIs is that these compounds inhibit one or more of theparasite’s aspartic proteases (termed plasmepsins), located on the food/digestive vacuole ornon-digestive vacuole [84–86].

Recently, we found that a non-nucleoside reverse transcriptase inhibitor, TMC-125, whichhad been shown to be highly active against HIV [87], demonstrated potent activity against

Grimberg and Mehlotra Page 7

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 8: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

P. falciparum [88]. It is theorized that this compound targets the parasite enzyme PfTERT, acatalytic reverse transcriptase component of telomerase, expressed in asexual blood-stageparasites that have begun DNA synthesis [89]. There appears to be only one gene for TERTin P. falciparum [89], P. vivax [90], and P. knowlesi [89], containing some highly conservedregions across species [89,90]. Since telomerase activity is likely to be necessary duringblood-stage parasite proliferation, it is hoped that screening other reverse transcriptaseinhibitors, or designing specific anti-telomerase compounds may lead to novel antimalarialdrugs with potent activity against multiple-species infections.

The potential for crossover between malaria and HIV treatments is undoubtedly intriguing,as most of the malaria-endemic areas also bear the brunt of the HIV pandemic. However, ata time when access to antiretroviral drugs is increasing, and new combinations ofantimalarial drugs, particularly ACTs, are being evaluated, it is important that potentialinteractions between therapies for these 2 infections are also understood [79,80,91]. In thisregard, it is important to mention that human drug-metabolizing enzymes CYP2B6 (phase Imetabolism) and UGT2B7 (phase II metabolism) play important roles in the metabolism ofantiretroviral drugs efavirenz, nevirapine, and zidovudine, as well as antimalarial artemisinindrugs, including their active metabolite DHA [92–94]. Functional polymorphisms inCYP2B6 and UGT2B7 are highly prevalent in the regions where HIV/AIDS and malaria co-occur [93–95]. Phenotypic consequences of polymorphisms in these enzyme genes on thepharmacokinetics and effectiveness of artemisinin drugs are yet to be determined, andcomprehensive studies evaluating the potential interactions between these antiretroviral andantimalarial drugs in individuals with or without polymorphisms are needed.

4.2.2. Drugs used for other diseases—Anti-cancer drugs—A new area inantimalarial drug discovery is the exploration of anti-cancer drugs that target cellularprograms such as cell proliferation, cell differentiation, and cell death [88,96,97]. Theinteresting revelation that artesunate is effective against certain types of cancer, and maytarget one or more of these cellular programs [98–100], also lends credence to this path ofinvestigation. Our recent study has shown that two anti-cancer compounds, SU-11274 andBay 43–9006, exhibit potent activities (IC50 values <1 μM) against P. falciparum [88]. Inhumans, SU-11274 acts as a specific inhibitor of the MET receptor tyrosine kinase activity[101]. Bay 43–9006, a dual-action inhibitor, targets the RAF/MEK/ERK signaling pathwayin tumor cells, and receptor tyrosine kinases VEGFR/PDGFR in tumor vasculature [102].The specific targets for SU-11274 and Bay 43–9006 activities against P. falciparum are notclear yet, but with the analysis of Plasmodium spp. genomes, plasmodial protein kinases(“Plasmodium kinome”) are emerging as highly promising targets for such compounds[103,104]. In addition, inhibition of protein kinases of the human host is being viewed asanother major parasiticidal strategy [97,103,105]. Recently, Doerig and colleagues [97]showed that the host PAK-MEK signaling pathway was selectively activated in P.falciparum-infected red blood cells. Selective inhibitors of PAK (IPA-3, IC50 value 2 μM)and MEK (U0126 [IC50 value 3 μM], PD184352 [IC50 value 7 μM]) inhibited the parasiteproliferation. Moreover, the MEK inhibitors showed in vitro parasiticidal effects onhepatocyte and erythrocyte stages of the rodent malaria parasite P. berghei, indicatingconservation of this subversive strategy in plasmodia [97].

As also discussed by Doerig and colleagues [97], several kinase-inhibiting drugs are nowused clinically, mostly in a variety of cancers, with many more in different stages of clinicaltrials. Evaluating these inhibitors for antimalarial properties would considerably reduce theoverall discovery/development cost and accelerate the process. A potential problemassociated with most of these inhibitors is toxicity, as they tend to have small therapeuticindices. Most fatalities in malaria occur in children under 5, which emphasizes the need fornot only highly efficacious but also very safe drugs. Although the kinase-inhibiting anti-

Grimberg and Mehlotra Page 8

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 9: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

cancer drugs have toxic effects, using them in malaria chemotherapy would require muchshorter treatment periods, hopefully limiting the problem of toxicity. It would also be ofinterest to see whether these drugs can be used in combination with artemisinin drugs, whichshow no “added toxicity”, and whether such a combination helps in reducing the effectiveparasiticidal concentration of these drugs, thus reducing their toxicity.

5. Next Generation of Antimalarial Treatment—Approaches and Candidates5.1. Development of New Drugs by Modifying Existing Compounds

5.1.1. Derivatization—One method to synthesize new antimalarial drugs is to start withthe chemical framework of existing antimalarial drugs; the 4- and 8-aminoquinolines andartemisinin drugs are the two common examples of this approach [106]. Recently, a varietyof 4-aminoquinoline derivatives (thiourea, triazine, and bisquinoline) have been found to behighly active against P. falciparum in vitro, and against P. yoelii or P. berghei in mice [107–109]. Synthesis/screening of 4-position modified quinoline-methanol compounds isanticipated to yield an efficacious and less toxic replacement for mefloquine [110–112]. Inaddition to 4-aminoquinoline analogs, extensive derivatization approaches followed bybetter understanding of structure-activity relationships and biotransformation mechanisms oftoxicity have also provided 8-aminoquinoline analogs with better pharmacologic andreduced toxicologic profiles [113].

Derivatives of artemisinin, particularly the water-soluble and efficacious artesunate, areanother obvious choice for the synthesis of new compounds [20,114]. Although highlypotent and fast acting, the currently available semisynthetic artemisinin derivatives are proneto hydrolysis, resulting in a short biological half-life and the generation of potentially toxicDHA [115,116]. Therefore, extensive efforts have been made to synthesize new derivativesof artemisinin and chemically-related compounds, which are more stable and exhibit potentin vitro [117,118] and in vivo [119–122] antimalarial activities. The in vitro activity of thesenew derivatives did not indicate any apparent cytotoxicity [118].

Thus, it appears that derivatization is one possible approach to prolong the clinicalusefulness of aminoquinoline and artemisinin classes of compounds. However, as a potentiallimitation of this approach, toxicity and cross-resistance with the parent compound may stilloccur in some of the derivatives [123].

5.1.2. Hybridization—Burgess et al. reported a highly innovative hybrid molecule thatcombines the pharmacophore of an active 4-amino-7-chloroquinoline antimalarial with aresistance-reversing group based on imipramine [124]. The compound was found to behighly active against both CQ-sensitive and CQ-resistant strains of P. falciparum in vitro,and against P. chabaudi in mice, with no obvious signs of toxicity. The authors named thisclass “reversed chloroquine (RCQ)”. Further studies showed that linking any of severalreversal-agent-like moieties to a 4-amino-7-chloroquinoline yielded good parasiticidalactivity [125]. Structure-activity relationship investigation indicated that RCQ moleculesinhibit hemozoin formation in the parasite’s digestive vacuole in a manner similar to that ofCQ [126].

Recently, in a deliberate rational design of antimalarials acting specifically on multipletargets, several hybrid molecules have been developed in what has been termed “covalentbitherapy” [127]. One interesting report described the synthesis of a novel artemisinin-quinine hybrid with potent antimalarial activity [128]. Artemisinin was reduced to DHA andcoupled to a carboxylic acid derivative of quinine via an ester linkage. This novel hybridmolecule had potent activity against CQ-sensitive and CQ-resistant strains of P. falciparumin vitro. The activity was superior to that of artemisinin alone, quinine alone, or a 1:1

Grimberg and Mehlotra Page 9

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 10: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

mixture of artemisinin and quinine [128]. Another example is a trioxane motif ofsesquiterpene lactone artemisinin covalently linked to a quinoline entity of anaminoquinoline (e.g., CQ), to form new modular molecules referred to as trioxaquines [127].The trioxaquines had more improved antimalarial activity than their individual components,indicating a potential additive/synergistic effect of the hybrids, as well as good oralbioavailability and low toxicity [127]. Such hybrid molecules have the potential to delay orcircumvent the development of resistance and reduce the risk of drug-drug adverseinteractions, the latter often seen with multiple-drug therapies. The antimalarial activities,combined with a good drug profile (preliminary absorption, metabolism, and safetyparameters) seem favorable for the selection of hybrid molecules for development as drugcandidates [129].

5.2. Finding New Antimalarial Compounds by Exploring Natural ProductsHistorically, drug discovery and development has greatly benefited from sourcingcompounds from nature. In fact, between 1981 and 2002, 61% of new chemical entitiesbrought to the market can be traced back to, or were inspired by, natural sources [130].Malaria drug discovery is no exception. The isolation of the antimalarial drug quinine fromcinchona bark was accomplished in 1820. The bark had long been used by indigenouspeoples in the Amazon region for the treatment of fevers. The Chinese herb qing hao(Artemisia annua) was also used as a treatment for fevers in China for more than 2,000years, but it was not until 1972 that the active compound artemisinin was extracted, and lateridentified as a potent antimalarial drug. The 1990s saw a demise in research into naturalproducts for drug discovery, due in part to the emergence of high-throughput screening andcombinatorial chemistry. Today, however, the current demand for novel compounds totackle emerging antimalarial resistance has stimulated new interest in their potential [131].Several screening projects utilizing different natural sources, from the rainforest to the deepsea and from exotic microorganisms to plants, have been carried out, resulting in severalinteresting antimalarial lead compounds with remarkable chemical diversity [132–136].Some such projects at the Medicines for Malaria Venture (MMV) are in the early-stagediscovery pipeline [46].

The mode of action of compounds originating from natural products is mostly unknown,and, in order to understand the basis for their pharmacological effects, research focused ontheir synergistic or antagonistic interactions is needed [137]. It is also clear that the muchdesired success of this approach faces several challenges, such as species selection criteria,screening procedures, pharmacological models and fractionation processes, as well asprediction of clinical safety and efficacy [138,139]. Nevertheless, it is hoped that once theactivity of natural medicinal products in humans is characterized, it can be used to identifynew molecular scaffolds which will form the basis of the next generation of antimalarialtherapies [140].

5.3. Finding New Antimalarial Drugs by Screening Diverse Chemical Libraries5.3.1. High-throughput screening—One of the major thrust areas for antimalarial drugdiscovery is to screen diverse chemical libraries using high-throughput assays. In recentyears, impressive efforts by several drug-screening groups at pharmaceutical companies andacademic institutions have generated a plethora of compounds, which may serve as newstarting points for potential antimalarial drugs with novel mechanisms. Here, we summarizefive such studies (Table 1).

i. From a screen of ~1.7 million compounds, Plouffe et al. [141] identified a diversecollection of ~6,000 small molecules comprised of >530 distinct scaffolds, all ofwhich showed potent antimalarial activity (<1.25 μM). Most known antimalarials

Grimberg and Mehlotra Page 10

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 11: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

were identified in this screen, thus validating their approach. In addition, theauthors identified many novel chemical scaffolds, which likely act through bothknown and novel pathways.

ii. Gamo et al. [142] screened nearly two million compounds in GlaxoSmithKline’schemical library for inhibitors of P. falciparum, of which 13,533 were confirmed toinhibit parasite growth by at least 80% at 2 μM concentration. More than 8,000 alsoshowed potent activity against the multidrug resistant strain Dd2. Further analysessuggested several novel mechanisms of their antimalarial action, such as inhibitionof protein kinases and host-pathogen interaction related targets. All of these provenplasmodial inhibitors, of which ~11,000 (82%) were previously proprietary andthus unknown to the general research community, were made public to acceleratethe pace of drug development for malaria.

iii. Guiguemde et al. [143] used a phenotypic forward chemical genetic approach toassay 309,474 chemicals to discover new antimalarial chemotypes. The primaryscreen resulted in ~1,300 hits with >80% activity against P. falciparum at 7 μM. Ofthese, antimalarial potencies of 172 compounds were re-confirmed to withintenfold by three laboratories using distinct methods providing the cross-validatedhit set. A reverse chemical genetic study of this set identified 19 new inhibitors offour validated drug targets, and 15 novel binders among 61 malarial proteins. Oneexemplar compound (SJ000025081) at 100 mg kg 1 b.i.d. × 3 days resulted in a90% suppression of P. yoelii parasitemia in mice.

iv. From a library of ~ 12,000 pure natural products and synthetic compounds withstructural features found in natural products, Rottmann et al. [144] identified 275primary hits with sub-micromolar activity against P. falciparum. After a series ofanalyses, a synthetic compound, related to the spiroazepineindole class, wasselected for a medicinal chemistry lead optimization effort. Synthesis andevaluation of about 200 derivatives yielded the spirotetrahydro-β-carboline (orspiroindolone) compound NITD609. This compound displayed potent activityagainst a panel of P. falciparum strains (average IC50 range 0.5 to 1.4 nM), with noevidence of diminished potency against drug-resistant strains. The IC50 valuesagainst a clone of multidrug resistant strain Dd2, however, increased 7- to 24-fold(attaining a mean of 3 to 11 nM) after 3 to 4 months of constant drug pressure.Subsequent passaging of drug-selected parasites in drug-free media for 4 monthsshowed no evidence of revertants, indicating that the resistance was stable. Theresistance was associated with point mutations in the gene encoding the P-typecation-transporter ATPase4 (pfatp4, PFL0590c). Further safety andpharmacological preclinical evaluation of NITD609 is currently ongoing to supportthe initiation of human clinical trials.

v. Using a high-throughput colorimetric assay for the detection of hemecrystallization inhibitors, Rush et al. [145] identified 17 hit compounds out of16,000 small molecules belonging to diverse structural classes. The IC50 values ofthese 17 hit compounds in the P. falciparum growth inhibition assay ranged from0.2 μM to 19 μM, and the compounds belonged to structurally related pyrimidineand 1,3-benzoxathiol-2-one classes. These hit compounds are being tested againstmultidrug-resistant strains of P. falciparum to further confirm that they do not showcross-resistance with currently deployed heme crystallization inhibitors.

5.4. Virtual ScreeningVirtual screening involves the rapid in silico assessment of large libraries of chemicalstructures in order to identify those which are most likely to bind to a drug target, typically a

Grimberg and Mehlotra Page 11

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 12: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

protein receptor or enzyme. This approach either alone or combined with high-throughputscreening is gaining popularity in the antimalarial drug discovery area. Plouffe et al. [141]showed that in some cases the mechanism of action of novel antimalarial compounds,identified by high-throughput cell-based screen, can be determined by in silico compoundactivity profiling. Using clustering by historical activities and the guilt-by-associationprinciple, the authors found that the compounds may segregate based on their mechanism ofaction (e.g., clusters of protein synthesis inhibitors or folic acid antagonists). Using an insilico approach, Woynarowski et al. [146] found that the targets for AT-specific DNA-reactive antitumor drugs adozelesin and bizelesin, which inhibited the growth of P.falciparum at picomolar concentrations, were “super-AT island” regions of the parasitegenome. Furthermore, investigation of the essentiality of a reaction in the metabolic networkof P. falciparum by deleting (knocking-out) such a reaction in silico [147], and comparisonof reconstructed metabolic network models from the parasite and its human host [148] hasrevealed essential enzymes from the parasite alone, representing new potential drug targets.As molecular databases of compounds and target structures are becoming larger, more andmore computational screening approaches are becoming available, providing new and betterinsights into in silico antimalarial drug discovery [149,150]. Using such computationalscreening approaches, novel and potent inhibitors of P. falciparum plasmepsins [151,152],glutathione-S-transferase [153], and DHFR [153] have been identified.

Thus, it is anticipated that with the aid of these new screening technologies, ongoing andnew projects, including those at the MMV, will keep the malaria drug discovery anddevelopment pipeline full with the next generation of potential malaria chemotherapeutics[46].

5.5. Genome-Based (“Targeted”) Drug DiscoveryThe complete genomes of the predominant malaria parasites P. falciparum and P. vivax, andthe human host are now known. The P. falciparum genome sequence comprises of 14chromosomes containing 5,300 identified genes [154]. Undoubtedly, the completion of thesePlasmodium spp. genomes, and their comparison with the human genome, provides theopportunity to discover parasite-specific, novel molecular targets for malaria therapy andprevention [155]. The information regarding gene expression and key regulatorycomponents of metabolism in P. falciparum is being assembled into various databases, togenerate and visualize network models of various cellular pathways and processes[156,157]. Efforts are also underway to characterize the importance of many of thesepathways and to predict their potential for pharmacological interventions [158–160].Furthermore, linking this increase in advancing new genetic information with the expressionof drug resistance will greatly assist in identifying sites of drug resistance as well asstrategies to improve the treatment [161].

A wealth of information on these themes has been presented in various recent articles [162–168]. The MMV currently has 30 projects in the discovery phase [46]. Out of these, 14discovery projects are on molecularly defined targets, with defined mechanisms of action,and 16 are on whole-cell activity, and therefore have unknown mechanisms of action. Inaddition to this, the MMV has a target database that currently contains more than 23 newtargets at different stages of screening and validation. The molecular targets in ongoing 14discovery projects at the MMV include P. falciparum cysteine proteases, termed falcipains[169–172], heat shock protein 90 (Hsp90) [173–175], DHFR with new series of inhibitors, avariety of kinases, histone deacetylases (HDAC), and dihydroorotate dehydrogenase(DHODH). Here, we present a summary of the recent key findings related to parasitekinases, HDAC, and DHODH as drug targets. As described below, since these enzymesseem essential for the parasite, they may be highly conserved among Plasmodium spp., andmutations in them are likely to be lethal. Therefore, the probability of the parasite becoming

Grimberg and Mehlotra Page 12

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 13: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

resistant to compounds directed against these enzymes should be minimal to none. Inaddition to these enzyme targets, we summarize key findings related to parasitesequestration receptors/anti-sequestration compounds as another interesting possible drugtarget.

5.5.1. Kinases—The life cycle of malaria parasites is very tightly regulated, since it passesthrough a succession of developmental stages that vary in terms of proliferation anddifferentiation. The successful completion of such a complex life cycle requires a strictcontrol of the cellular machinery that carries out phosphorylation, transcriptional control,post-transcriptional control, and protein degradation. These mechanisms most likely involvefine interactions among various cell-cycle proteins (particularly kinases), which mayrepresent strategic targets for pharmacological interventions [96,104,176].

There are a large number and variety of kinases (“Plasmodium kinome”), many of which areclearly distinct from human protein kinases, and thus are considered as targets of choice forantimalarial drug development [176,177]. A serine/threonine protein kinase, casein kinase2α (PfCK2α), crucial for asexual blood-stage parasites, has been identified as a potentialtarget for antimalarial chemotherapeutic intervention [178]. Among cyclin-dependentprotein kinases, Pfmrk is the most attractive target for antimalarial drug development; avariety of inhibitors selectively inhibit Pfmrk at sub-micromolar concentrations [179–181].Calcium-dependent protein kinase 1 (PfCDPK1), essential for parasite survival, can beinhibited by small-molecule compounds at nanomolar concentrations [182,183]. An orphanprotein kinase PfPK7, significant for asexual stage development in humans and oocystproduction in mosquitoes, has recently been identified as a lead target [184–187]. Inaddition, mitogen-activated protein kinase 2 (PfMAP-2), essential for completion of theasexual cycle 188, NIMA-related protein kinase Pfnek-2, predominantly expressed in, andrequired for transmission of, gametocytes [189], and cAMP-dependent protein kinase(PfPKA), essential for parasite growth and survival [190], hold the promise of beingselective and effective targets for the development of new antimalarial drugs.

5.5.2. Histone deacetylases—Histone acetylation plays key roles in regulating genetranscription in both prokaryotes and eukaryotes, the acetylated form inducing geneexpression while deacetylation silences genes. A number of HDAC inhibitors are currentlyin clinical trials, alone or in combination, for the treatment of a variety of cancers, parasitic/infectious diseases, and hemoglobinopathies [191,192]. Plasmodium falciparum has at leastfive putative HDACs, which play a major role in transcriptional regulation of the parasitelife cycle [193]. Among these, PfHDAC1 in particular is considered as a promising newantimalarial drug target. A variety of PfHDAC1 inhibitors showed potent in vitroantimalarial activity (IC50 values in nM range) against CQ-sensitive and CQ-resistant strainsof the parasite, and several of the inhibitors were significantly more toxic to the parasitesthan to mammalian cells [194,195]. Furthermore, an HDAC inhibitor WR301801 (YC-2–88)exhibited cures in P. berghei-infected mice at a dose of 52 mg/kg/day orally, whencombined with subcurative doses of CQ [196]. Recently, using a modified schizontmaturation assay, certain HDAC inhibitors were found to exert potent activity (at nMconcentrations) against multidrug-resistant clinical isolates of both P. falciparum (n = 24)and P. vivax (n = 25) from Papua, Indonesia, suggesting that these inhibitors may bepromising candidates for antimalarial therapy in geographical locations where both speciesare endemic [197].

5.5.3. Dihydroorotate dehydrogenase—Erythrocytic stages of P. falciparum seem tomaintain an active mitochondrial electron transport chain to serve just one metabolicfunction: regeneration of ubiquinone required as the electron acceptor for DHODH, anessential enzyme for de novo pyrimidine biosynthesis [198]. For this very reason,

Grimberg and Mehlotra Page 13

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 14: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

PfDHODH is also receiving increasing attention as a new target for antimalarial drugdiscovery [199,200]. A variety of potent inhibitors of PfDHODH showing strong selectivityfor the malarial enzyme over that from the human host have been designed and synthesized,and identified by high-throughput screening [201–204]. Potent activity against Plasmodiumspp. parasites in vitro at nM concentrations, with good correlation between activity againstthe parasites and the enzyme from the parasites has been observed for a number of theseinhibitors [201,205]. Lead optimization of a triazolopyrimidine-based series of inhibitors hasidentified Genz-667348, which is orally bioavailable, with prolonged plasma exposure, andcures P. berghei infection in mice at a total dose of 100 mg/kg/day [201]. Further refinementof the structure-activity relationship within this series is underway, with 2 recent analogs ofGenz-667348 undergoing further testing to determine if they represent suitable candidatesfor preclinical development [201].

5.5.4. Anti-sequestration compounds—An interesting possibility—In addition,new compounds may participate in the combat against falciparum malaria not by directlykilling but preventing the parasite from causing severe forms of the disease. One way toachieve this is to inhibit the parasite’s ability to sequester. Sequestration occurs becauseparasite-derived adhesins, expressed on the surface of mature infected erythrocytes, bind toreceptors on human cells (often referred to simply as cytoadherence or cytoadhesion).Organ-specific sequestration in brain and placenta play an important role in the pathogenesisof cerebral and placental malaria, respectively [5,206]. Plasmodium falciparum-infectederythrocytes have been shown to have the potential for binding to a diverse array ofendothelial receptors; however, only two of these receptors, CD36 and intracellular adhesionmolecule 1 (ICAM1), have been studied in detail [206]. Chondroitin sulfate A (CSA) hasbeen consistently identified as the dominant placental adhesion receptor [5].

In recent years, some progress has been made in inhibiting cytoadherence that is mediatedby these receptors. CD36-mediated cytoadherence can be inhibited in vitro by antiretroviralPIs ritonavir and saquinavir [207], and polysaccharides derived from seaweed(carrageenans) [208]. A randomized clinical trial of Thai patients with uncomplicatedmalaria showed that levamisole, an antihelminth drug, known to block cytoadherence toCD36, when used as an adjunctive therapy with quinine and doxycycline, resulted inincreased numbers of early-mid trophozoites in the peripheral blood [209]. It was suggestedthat levamisole prevented the sequestration of these parasites as they matured from ringstages following treatment [209]. Using the crystal structure of ICAM1, Dormeyer andcolleagues [210] found that (+)-epigalloylcatechin gallate, a naturally occurring polyphenolcompound in green tea, inhibited cytoadherence to ICAM1 in a dose-dependent manner withtwo variant ICAM 1-binding parasite lines. Using CSA-expressing CHO-K1 cells and exvivo human placental tissue, Andrews et al. [211] showed that two carrageenans and acellulose sulfate (CS10) were able to inhibit cytoadhesion to CHO-K1 cells as well ashuman placental tissue; the inhibitory effect on CHO-K1 cells was dose-dependent.

Inhibition of cell adhesion processes is becoming increasingly interesting in the discovery ofnovel therapeutics [212]. Although our understanding of the molecular mechanisms ofparasite adhesion is still incomplete, and many questions remain unanswered [206], thediscoveries outlined above open up the possibility of developing therapeutic interventionsaimed at blocking or reversing parasite adhesion.

6. A Final NoteIn these various approaches to: (i) efficient use of existing as well as forthcoming newantimalarial drugs and (ii) discovery of novel antimalarial drugs, one important aspect notdiscussed here is the pharmacology of antimalarial agents. Since malaria is a blood

Grimberg and Mehlotra Page 14

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 15: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

infection, the response to an antimalarial treatment is likely determined by the concentrationof a drug and/or its active metabolite(s) in the blood. Although antimalarial drugs have beenin use since the 1930s, until recently, no data were available on the absorption-distribution-metabolism-excretion parameters of these drugs in humans. Over the past 10–15 years, therehave been many methodological developments in the area of pharmacology related toantimalarials. Through these advancements, pharmacokinetics and the major metabolicpathways of a number of antimalarial drugs have been elucidated [33,167,213]. However,little emphasis has been placed on understanding the basic mechanisms responsible for thepharmacokinetic and pharmacodynamic behaviors of the major classes of antimalarial drugsin various populations. Particularly, what role does human genetic diversity play in theoutcomes of treatment, such as failure, resistance, and side effects, with antimalarial drugs?Since drug combinations are increasingly being considered to successfully treat malariaworldwide, such information is essential to optimize antimalarial drug regimens, particularlynew regimens, in order to achieve high treatment effectiveness across all malaria-endemicregions.

7. ConclusionsIn the battle against malaria, we are currently at a stalemate at best. If and when theresistance to artemisinin drugs further develops and spreads, we will be caught unarmed,without any effective weapon against this deadly parasite. Looking at the antimalarial drugdevelopment pipeline, new classes of agents, and, in the meantime, a re-optimization ofexisting drugs using new creative strategies are needed. Re-optimization of existing drugsthrough replacement/rotation and combination approaches may prolong their life spans foreffective treatment and prophylaxis, until new treatments are found. The future ofantimalarial drug discovery lies in innovative thinking and novel areas, some currently underexploration and some yet to be explored. As described in this article, there are a number ofdifferent promising approaches to the identification of new antimalarials. Among theseapproaches, repurposing drugs that are used for other infections or diseases, exploringnatural products with medicinal significance (in malaria), and target-based drug discoveryare of particular interest. In addition, evolving technologies, such as high-throughput cell-based assays and virtual screening, will certainly be useful in discovering new effectivetreatments for malaria relatively quickly. We do not have any choice but to explore many orall of these approaches in parallel, until existing drugs with renewed potential, as well asentirely new series of compounds are available for deployment in the field. The discovery ofnew treatments for malaria, along with their proper execution in the field, will contribute toan important achievement of controlling, and eventually eradicating, this global infectiousdisease.

AcknowledgmentsThe authors thank Kerry O’Connor Grimberg, Dave McNamara, Kathy Andrews, and Cara Henry-Halldin, forhelpful discussions and critical evaluation of this manuscript. RKM dedicates this article to the loving memories ofhis mentors late B. N. Singh and O. P. Shukla, from whom he learnt philosophy of Science and realities of Life.RKM is also thankful to his teacher Sai Baba, whose unconditional love helps him think, write, and beyond. BTG issupported by grants from; the National Institute of Health (AI079388), The International Society for Advancementof Cytometry Scholars Program, and the Case Western Reserve University Vision Fund.

References1. Bates I, Fenton C, Gruber J, Lalloo D, Medina Lara A, Squire SB, Theobald S, Thomson R,

Tolhurst R. Vulnerability to malaria, tuberculosis, and HIV/AIDS infection and disease. Part 1:Determinants operating at individual and household level. Lancet Infect Dis. 2004; 4:267–277.[PubMed: 15120343]

Grimberg and Mehlotra Page 15

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 16: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

2. Stratton L, O’Neill MS, Kruk ME, Bell ML. The persistent problem of malaria: Addressing thefundamental causes of a global killer. Soc Sci Med. 2008; 67:854–862. [PubMed: 18583009]

3. WHO. World Malaria Report. 2010.http://www.who.int/malaria/world_malaria_report_2010/en/index.html/

4. Roca-Feltrer A, Carneiro I, Armstrong Schellenberg JR. Estimates of the burden of malariamorbidity in Africa in children under the age of 5 years. Trop Med Int Health. 2008; 13:771–783.[PubMed: 18363586]

5. Rogerson SJ. Malaria in pregnancy and the newborn. Adv Exp Med Biol. 2010; 659:139–152.[PubMed: 20204762]

6. Schantz-Dunn J, Nour NM. Malaria and pregnancy: A global health perspective. Rev ObstetGynecol. 2009; 2:186–192. [PubMed: 19826576]

7. Krogstad, DJ. Malaria. In: Guerrant, RL.; Walker, DH.; Weller, PF., editors. Tropical InfectiousDiseases-Principles, Pathogens, & Practice. Vol. 1. Churchill Livingstone; Philadelphia, PA, USA:1999. p. 736-766.

8. Service, M.; Townson, H. The anopheles vector. In: Gilles, H.; Warrell, DA., editors. EssentialMalariology. Arnold; London, UK: 2002. p. 59-84.

9. Gurarie D, Zimmerman PA, King CH. Dynamic regulation of single- and mixed-species malariainfection: Insights to specific and non-specific mechanisms of control. J Theor Biol. 2006; 240:185–199. [PubMed: 16263133]

10. Mayxay M, Pukrittayakamee S, Newton PN, White NJ. Mixed-species malaria infections inhumans. Trends Parasitol. 2004; 20:233–240. [PubMed: 15105024]

11. Zimmerman PA, Mehlotra RK, Kasehagen LJ, Kazura JW. Why do we need to know more aboutmixed Plasmodium species infections in humans? Trends Parasitol. 2004; 20:440–447. [PubMed:15324735]

12. Baird JK. Neglect of Plasmodium vivax malaria. Trends Parasitol. 2007; 23:533–539. [PubMed:17933585]

13. Price RN, Tjitra E, Guerra CA, Yeung S, White NJ, Anstey NM. Vivax malaria: Neglected and notbenign. Am J Trop Med Hyg. 2007; 77(Suppl 6):9–87.

14. Greenwood BM, Fidock DA, Kyle DE, Kappe SH, Alonso PL, Collins FH, Duffy PE. Malaria:Progress, perils, and prospects for eradication. J Clin Invest. 2008; 118:1266–1276. [PubMed:18382739]

15. Hemingway J, Ranson H. Insecticide resistance in insect vectors of human disease. Annu RevEntomol. 2000; 45:371–391. [PubMed: 10761582]

16. Girard MP, Reed ZH, Friede M, Kieny MP. A review of human vaccine research and development:Malaria. Vaccine. 2007; 25:1567–1580. [PubMed: 17045367]

17. Schlitzer M. Antimalarial drugs—What is in use and what is in the pipeline. Arch Pharm(Weinheim). 2008; 341:149–163. [PubMed: 18297679]

18. White NJ. Qinghaosu (artemisinin): The price of success. Science. 2008; 320:330–334. [PubMed:18420924]

19. Wernsdorfer WH, Payne D. The dynamics of drug resistance in Plasmodium falciparum.Pharmacol Ther. 1991; 50:95–121. [PubMed: 1891480]

20. Wiesner J, Ortmann R, Jomaa H, Schlitzer M. New antimalarial drugs. Angew Chem Int Ed Engl.2003; 42:5274–5293. [PubMed: 14613157]

21. Croft AM. A lesson learnt: The rise and fall of Lariam and Halfan. JR Soc Med. 2007; 100:170–174.

22. Luzzatto L. The rise and fall of the antimalarial Lapdap: A lesson in pharmacogenetics. Lancet.2010; 376:739–741. [PubMed: 20599264]

23. WHO. The use of antimalarial drugs: Report of an informal consultation (WHO/CDS/RBM/2001.33). http://rbm.who.int/cmc_upload/0/000/014/923/am_toc.htm/

24. Bosman A, Mendis KN. A major transition in malaria treatment: The adoption and deployment ofartemisinin-based combination therapies. Am J Trop Med Hyg. 2007; 77(Suppl 6):93–197.

25. Hastings I. How artemisinin-containing combination therapies slow the spread of antimalarial drugresistance. Trends Parasitol. 201010.1016/j.pt.2010.09.005

Grimberg and Mehlotra Page 16

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 17: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

26. Musset L, Bouchaud O, Matheron S, Massias L, Le Bras J. Clinical atovaquone-proguanilresistance of Plasmodium falciparum associated with cytochrome b codon 268 mutations.Microbes Infect. 2006; 8:2599–2604. [PubMed: 16962361]

27. Hyde JE. Drug-resistant malaria. Trends Parasitol. 2005; 21:494–498. [PubMed: 16140578]28. Hayton K, Su XZ. Genetic and biochemical aspects of drug resistance in malaria parasites. Curr

Drug Targets Infect Disord. 2004; 4:1–10. [PubMed: 15032630]29. Hyde JE. Drug-resistant malaria—An insight. FEBS J. 2007; 274:4688–4698. [PubMed:

17824955]30. Ekland EH, Fidock DA. Advances in understanding the genetic basis of antimalarial drug

resistance. Curr Opin Microbiol. 2007; 10:363–370. [PubMed: 17709280]31. Kidgell C, Winzeler EA. Using the genome to dissect the molecular basis of drug resistance.

Future Microbiol. 2006; 1:185–199. [PubMed: 17661664]32. Valderramos SG, Fidock DA. Transporters involved in resistance to antimalarial drugs. Trends

Pharmacol Sci. 2006; 27:594–601. [PubMed: 16996622]33. Mehlotra RK, Henry-Halldin CN, Zimmerman PA. Application of pharmacogenomics to malaria:

A holistic approach for successful chemotherapy. Pharmacogenomics. 2009; 10:435–449.[PubMed: 19290792]

34. Meshnick SR, Taylor TE, Kamchonwongpaisan S. Artemisinin and the antimalarialendoperoxides: From herbal remedy to targeted chemotherapy. Microbiol Rev. 1996; 60:301–315.[PubMed: 8801435]

35. Teuscher F, Gatton ML, Chen N, Peters J, Kyle DE, Cheng Q. Artemisinin-induced dormancy inPlasmodium falciparum: Duration, recovery rates, and implications in treatment failure. J InfectDis. 2010; 202:1362–1368. [PubMed: 20863228]

36. Witkowski B, Lelievre J, Barragan MJ, Laurent V, Su XZ, Berry A, Benoit-Vical F. Increasedtolerance to artemisinin in Plasmodium falciparum is mediated by a quiescence mechanism.Antimicrob Agents Chemother. 2010; 54:1872–1877. [PubMed: 20160056]

37. Enserink M. Malaria’s drug miracle in danger. Science. 2010; 328:844–846. [PubMed: 20466917]38. Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, Lwin KM, Ariey F, Hanpithakpong W,

Lee SJ, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–467. [PubMed: 19641202]

39. Noedl H, Se Y, Schaecher K, Smith BL, Socheat D, Fukuda MM. Evidence of artemisinin-resistantmalaria in western Cambodia. N Engl J Med. 2008; 359:2619–2620. [PubMed: 19064625]

40. Anderson TJ, Nair S, Nkhoma S, Williams JT, Imwong M, Yi P, Socheat D, Das D, ChotivanichK, Day NP, et al. High heritability of malaria parasite clearance rate indicates a genetic basis forartemisinin resistance in western Cambodia. J Infect Dis. 2010; 201:1326–1330. [PubMed:20350192]

41. Cui L, Su XZ. Discovery, mechanisms of action and combination therapy of artemisinin. ExpertRev Anti-Infect Ther. 2009; 7:999–1013. [PubMed: 19803708]

42. O’Neill PM, Barton VE, Ward SA. The molecular mechanism of action of artemisinin—Thedebate continues. Molecules. 2010; 15:1705–1721. [PubMed: 20336009]

43. Eckstein-Ludwig U, Webb RJ, Van Goethem ID, East JM, Lee AG, Kimura M, O’Neill PM, BrayPG, Ward SA, Krishna S. Artemisinins target the SERCA of Plasmodium falciparum. Nature.2003; 424:957–961. [PubMed: 12931192]

44. Imwong M, Dondorp AM, Nosten F, Yi P, Mungthin M, Hanchana S, Das D, Phyo AP, Lwin KM,Pukrittayakamee S, et al. Exploring the contribution of candidate genes to artemisinin resistance inPlasmodium falciparum. Antimicrob Agents Chemother. 2010; 54:2886–2892. [PubMed:20421395]

45. Dondorp AM, Yeung S, White L, Nguon C, Day NP, Socheat D, von Seidlein L. Artemisininresistance: Current status and scenarios for containment. Nat Rev Microbiol. 2010; 8:272–280.[PubMed: 20208550]

46. Olliaro P, Wells TN. The global portfolio of new antimalarial medicines under development. ClinPharmacol Ther. 2009; 85:584–595. [PubMed: 19404247]

Grimberg and Mehlotra Page 17

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 18: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

47. Laufer MK, Thesing PC, Eddington ND, Masonga R, Dzinjalamala FK, Takala SL, Taylor TE,Plowe CV. Return of chloroquine antimalarial efficacy in Malawi. N Engl J Med. 2006;355:1959–1966. [PubMed: 17093247]

48. Zhou Z, Griffing SM, de Oliveira AM, McCollum AM, Quezada WM, Arrospide N, Escalante AA,Udhayakumar V. Decline in sulfadoxine-pyrimethamine-resistant alleles after change in drugpolicy in the Amazon region of Peru. Antimicrob Agents Chemother. 2008; 52:739–741.[PubMed: 18025120]

49. Walliker D, Hunt P, Babiker H. Fitness of drug-resistant malaria parasites. Acta Trop. 2005;94:251–259. [PubMed: 15845348]

50. Kublin JG, Cortese JF, Njunju EM, Mukadam RA, Wirima JJ, Kazembe PN, Djimde AA, KouribaB, Taylor TE, Plowe CV. Reemergence of chloroquine-sensitive Plasmodium falciparum malariaafter cessation of chloroquine use in Malawi. J Infect Dis. 2003; 187:1870–1875. [PubMed:12792863]

51. Laufer MK, Takala-Harrison S, Dzinjalamala FK, Stine OC, Taylor TE, Plowe CV. Return ofchloroquine-susceptible falciparum malaria in Malawi was a reexpansion of diverse susceptibleparasites. J Infect Dis. 2010; 202:801–808. [PubMed: 20662717]

52. Mita T, Kaneko A, Lum JK, Zungu IL, Tsukahara T, Eto H, Kobayakawa T, Bjorkman A, TanabeK. Expansion of wild type allele rather than back mutation in pfcrt explains the recent recovery ofchloroquine sensitivity of Plasmodium falciparum in Malawi. Mol Biochem Parasitol. 2004;135:159–163. [PubMed: 15287598]

53. Mwai L, Ochong E, Abdirahman A, Kiara SM, Ward S, Kokwaro G, Sasi P, Marsh K, BorrmannS, Mackinnon M, et al. Chloroquine resistance before and after its withdrawal in Kenya. Malar J.2009; 8:106. [PubMed: 19450282]

54. Chen N, Gao Q, Wang S, Wang G, Gatton M, Cheng Q. No genetic bottleneck in Plasmodiumfalciparum wild-type Pfcrt alleles reemerging in Hainan Island, China, following high-levelchloroquine resistance. Antimicrob Agents Chemother. 2008; 52:345–347. [PubMed: 17954685]

55. Wang X, Mu J, Li G, Chen P, Guo X, Fu L, Chen L, Su X, Wellems TE. Decreased prevalence ofthe Plasmodium falciparum chloroquine resistance transporter 76T marker associated withcessation of chloroquine use against P. falciparum malaria in Hainan, People’s Republic of China.Am J Trop Med Hyg. 2005; 72:410–414. [PubMed: 15827277]

56. McCollum AM, Mueller K, Villegas L, Udhayakumar V, Escalante AA. Common origin andfixation of Plasmodium falciparum dhfr and dhps mutations associated with sulfadoxine-pyrimethamine resistance in a low-transmission area in South America. Antimicrob AgentsChemother. 2007; 51:2085–2091. [PubMed: 17283199]

57. Khim N, Bouchier C, Ekala MT, Incardona S, Lim P, Legrand E, Jambou R, Doung S, PuijalonOM, Fandeur T. Countrywide survey shows very high prevalence of Plasmodium falciparummultilocus resistance genotypes in Cambodia. Antimicrob Agents Chemother. 2005; 49:3147–3152. [PubMed: 16048916]

58. Ginsburg H. Should chloroquine be laid to rest? Acta Trop. 2005; 96:16–23. [PubMed: 16054105]59. Martin SK, Oduola AM, Milhous WK. Reversal of chloroquine resistance in Plasmodium

falciparum by verapamil. Science. 1987; 235:899–901. [PubMed: 3544220]60. Egan TJ, Kaschula CH. Strategies to reverse drug resistance in malaria. Curr Opin Infect Dis.

2007; 20:598–604. [PubMed: 17975410]61. Guantai E, Chibale K. Chloroquine resistance: Proposed mechanisms and countermeasures. Curr

Drug Deliv. 2010; 7:312–323. [PubMed: 20695842]62. Henry M, Alibert S, Orlandi-Pradines E, Bogreau H, Fusai T, Rogier C, Barbe J, Pradines B.

Chloroquine resistance reversal agents as promising antimalarial drugs. Curr Drug Targets. 2006;7:935–948. [PubMed: 16918322]

63. van Schalkwyk DA, Egan TJ. Quinoline-resistance reversing agents for the malaria parasitePlasmodium falciparum. Drug Resist Updat. 2006; 9:211–226. [PubMed: 17064951]

64. Ward SA, Bray PG. Is reversal of chloroquine resistance ready for the clinic? Lancet. 2001;357:904. [PubMed: 11289344]

65. Sendagire H, Kaddumukasa M, Ndagire D, Aguttu C, Nassejje M, Pettersson M, Swedberg G,Kironde F. Rapid increase in resistance of Plasmodium falciparum to chloroquine-Fansidar in

Grimberg and Mehlotra Page 18

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 19: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

Uganda and the potential of amodiaquine-Fansidar as a better alternative. Acta Trop. 2005;95:172–182. [PubMed: 16018959]

66. Wootton DG, Opara H, Biagini GA, Kanjala MK, Duparc S, Kirby PL, Woessner M, Neate C,Nyirenda M, Blencowe H, et al. Open-label comparative clinical study of chlorproguanil-dapsonefixed dose combination (Lapdap) alone or with three different doses of artesunate foruncomplicated Plasmodium falciparum malaria. PLoS One. 2008; 3:e1779. [PubMed: 18320064]

67. Price RN, Dorsey G, Ashley EA, Barnes KI, Baird JK, d’Alessandro U, Guerin PJ, Laufer MK,Naidoo I, Nosten F, et al. World Antimalarial Resistance Network I: Clinical efficacy ofantimalarial drugs. Malar J. 2007; 6:119. [PubMed: 17822532]

68. Schlagenhauf P, Adamcova M, Regep L, Schaerer MT, Rhein HG. The position of mefloquine as a21st century malaria chemoprophylaxis. Malar J. 2010; 9:357. [PubMed: 21143906]

69. Dow G, Bauman R, Caridha D, Cabezas M, Du F, Gomez-Lobo R, Park M, Smith K, Cannard K.Mefloquine induces dose-related neurological effects in a rat model. Antimicrob AgentsChemother. 2006; 50:1045–1053. [PubMed: 16495267]

70. Dow GS, Caridha D, Goldberg M, Wolf L, Koenig ML, Yourick DL, Wang Z. Transcriptionalprofiling of mefloquine-induced disruption of calcium homeostasis in neurons in vitro. Genomics.2005; 86:539–550. [PubMed: 16109470]

71. Nevin RL, Pietrusiak PP, Caci JB. Prevalence of contraindications to mefloquine use among USAmilitary personnel deployed to Afghanistan. Malar J. 2008; 7:30. [PubMed: 18267019]

72. DoD. Policy memorandum on the use of mefloquine (LariamR) in malaria prophylaxis.http://www.lariaminfo.org/pdfs/policy-memo-secy-defense%20malaria-prophylaxis.pdf

73. Shanks GD, Oloo AJ, Aleman GM, Ohrt C, Klotz FW, Braitman D, Horton J, Bruekner R. A newprimaquine analogue, tafenoquine (WR 238605), for prophylaxis against Plasmodium falciparummalaria. Clin Infect Dis. 2001; 33:1968–1974. [PubMed: 11700577]

74. Walsh DS, Looareesuwan S, Wilairatana P, Heppner DG Jr, Tang DB, Brewer TG, ChokejindachaiW, Viriyavejakul P, Kyle DE, Milhous WK, et al. Randomized dose-ranging study of the safetyand efficacy of WR 238605 (Tafenoquine) in the prevention of relapse of Plasmodium vivaxmalaria in Thailand. J Infect Dis. 1999; 180:1282–1287. [PubMed: 10479159]

75. Dutta GP, Puri SK. New antimalarial drug development in india: radical curative agents CDRI80/53 (Elubaquine) and WR 238605 (Tafenoquine). Proc Indian Nat Sci Acad. 2003; B69:871–882.

76. Crockett M, Kain KC. Tafenoquine: A promising new antimalarial agent. Exp Opin Invest Drugs.2007; 16:705–715.

77. Elmes NJ, Nasveld PE, Kitchener SJ, Kocisko DA, Edstein MD. The efficacy and tolerability ofthree different regimens of tafenoquine versus primaquine for post-exposure prophylaxis ofPlasmodium vivax malaria in the Southwest Pacific. Trans R Soc Trop Med Hyg. 2008; 102:1095–1101. [PubMed: 18541280]

78. Nasveld PE, Edstein MD, Reid M, Brennan L, Harris IE, Kitchener SJ, Leggat PA, Pickford P,Kerr C, Ohrt C, et al. Randomized, double-blind study of the safety, tolerability, and efficacy oftafenoquine versus mefloquine for malaria prophylaxis in nonimmune subjects. AntimicrobAgents Chemother. 2010; 54:792–798. [PubMed: 19995933]

79. Khoo S, Back D, Winstanley P. The potential for interactions between antimalarial andantiretroviral drugs. Aids. 2005; 19:995–1005. [PubMed: 15958830]

80. Skinner-Adams TS, McCarthy JS, Gardiner DL, Andrews KT. HIV and malaria co-infection:Interactions and consequences of chemotherapy. Trends Parasitol. 2008; 24:264–271. [PubMed:18456554]

81. Skinner-Adams TS, McCarthy JS, Gardiner DL, Hilton PM, Andrews KT. Antiretrovirals asantimalarial agents. J Infect Dis. 2004; 190:1998–2000. [PubMed: 15529265]

82. Parikh S, Gut J, Istvan E, Goldberg DE, Havlir DV, Rosenthal PJ. Antimalarial activity of humanimmunodeficiency virus type 1 protease inhibitors. Antimicrob Agents Chemother. 2005;49:2983–2985. [PubMed: 15980379]

83. Peatey CL, Andrews KT, Eickel N, MacDonald T, Butterworth AS, Trenholme KR, Gardiner DL,McCarthy JS, Skinner-Adams TS. Antimalarial asexual stage-specific and gametocytocidal

Grimberg and Mehlotra Page 19

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 20: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

activities of HIV protease inhibitors. Antimicrob Agents Chemother. 2010; 54:1334–1337.[PubMed: 20028821]

84. Andrews KT, Fairlie DP, Madala PK, Ray J, Wyatt DM, Hilton PM, Melville LA, Beattie L,Gardiner DL, Reid RC, et al. Potencies of human immunodeficiency virus protease inhibitors invitro against Plasmodium falciparum and in vivo against murine malaria. Antimicrob AgentsChemother. 2006; 50:639–648. [PubMed: 16436721]

85. Parikh S, Liu J, Sijwali P, Gut J, Goldberg DE, Rosenthal PJ. Antimalarial effects of humanimmunodeficiency virus type 1 protease inhibitors differ from those of the aspartic proteaseinhibitor pepstatin. Antimicrob Agents Chemother. 2006; 50:2207–2209. [PubMed: 16723585]

86. Skinner-Adams TS, Andrews KT, Melville L, McCarthy J, Gardiner DL. Synergistic interactionsof the antiretroviral protease inhibitors saquinavir and ritonavir with chloroquine and mefloquineagainst Plasmodium falciparum in vitro. Antimicrob Agents Chemother. 2007; 51:759–762.[PubMed: 17088482]

87. Janssen PA, Lewi PJ, Arnold E, Daeyaert F, de Jonge M, Heeres J, Koymans L, Vinkers M,Guillemont J, Pasquier E, et al. In search of a novel anti-HIV drug: multidisciplinary coordinationin the discovery of 4-[[4-[[4-[(1E)-2-cyanoethenyl]-2,6-dimethylphenyl]amino]-2-pyrimidinyl]amino]benzonitrile (R278474, rilpivirine). J Med Chem. 2005; 48:1901–1909.[PubMed: 15771434]

88. Grimberg BT, Jaworska MM, Hough LB, Zimmerman PA, Phillips JG. Addressing the malariadrug resistance challenge using flow cytometry to discover new antimalarials. Bioorg Med ChemLett. 2009; 19:5452–5457. [PubMed: 19666223]

89. Figueiredo LM, Rocha EP, Mancio-Silva L, Prevost C, Hernandez-Verdun D, Scherf A. Theunusually large Plasmodium telomerase reverse-transcriptase localizes in a discrete compartmentassociated with the nucleolus. Nucl Acid Res. 2005; 33:1111–1122.

90. Durand PM, Oelofse AJ, Coetzer TL. An analysis of mobile genetic elements in three Plasmodiumspecies and their potential impact on the nucleotide composition of the P. falciparum genome.BMC Genomics. 2006; 7:282. [PubMed: 17083741]

91. Dooley KE, Flexner C, Andrade AS. Drug interactions involving combination antiretroviraltherapy and other anti-infective agents: Repercussions for resource-limited countries. J Infect Dis.2008; 198:948–961. [PubMed: 18713054]

92. Belanger AS, Caron P, Harvey M, Zimmerman PA, Mehlotra RK, Guillemette C. Glucuronidationof the antiretroviral drug efavirenz by UGT2B7 and an in vitro investigation of drug-druginteraction with zidovudine. Drug Metab Dispos. 2009; 37:1793–1796. [PubMed: 19487252]

93. Mehlotra RK, Bockarie MJ, Zimmerman PA. Prevalence of UGT1A9 and UGT2B7nonsynonymous single nucleotide polymorphisms in West African, Papua New Guinean, andNorth American populations. Eur J Clin Pharmacol. 2007; 63:1–8. [PubMed: 17115150]

94. Mehlotra RK, Ziats MN, Bockarie MJ, Zimmerman PA. Prevalence of CYP2B6 alleles in malaria-endemic populations of West Africa and Papua New Guinea. Eur J Clin Pharmacol. 2006; 62:267–275. [PubMed: 16506047]

95. Mehlotra RK, Bockarie MJ, Zimmerman PA. CYP2B6 983T>C polymorphism is prevalent in WestAfrica but absent in Papua New Guinea: Implications for HIV/AIDS treatment. Br J ClinPharmacol. 2007; 64:391–395. [PubMed: 17391322]

96. Kozlov S, Waters NC, Chavchich M. Leveraging cell cycle analysis in anticancer drug discoveryto identify novel plasmodial drug targets. Infect Disord Drug Targets. 2010; 10:165–190.[PubMed: 20334621]

97. Sicard A, Semblat JP, Doerig C, Hamelin R, Moniatte M, Dorin-Semblat D, Spicer JA, SrivastavaA, Retzlaff S, Heussler V, et al. Activation of a PAK-MEK signalling pathway in malaria parasite-infected erythrocytes. Cell Microbiol. 201010.1111/j.1462–5822.2011.01582.x

98. Firestone GL, Sundar SN. Anticancer activities of artemisinin and its bioactive derivatives. ExpertRev Mol Med. 2009; 11:e32. [PubMed: 19883518]

99. Ghantous A, Gali-Muhtasib H, Vuorela H, Saliba NA, Darwiche N. What made sesquiterpenelactones reach cancer clinical trials? Drug Discov Today. 2010; 15:668–678. [PubMed: 20541036]

100. Li-Weber M. Targeting apoptosis pathways in cancer by Chinese medicine. Cancer Lett. 2010Epub ahead of print.

Grimberg and Mehlotra Page 20

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 21: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

101. Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K,Christensen JG, et al. A novel small molecule met inhibitor induces apoptosis in cellstransformed by the oncogenic TPR-MET tyrosine kinase. Cancer Res. 2003; 63:5462–5469.[PubMed: 14500382]

102. Adnane L, Trail PA, Taylor I, Wilhelm SM. Sorafenib (BAY 43–9006, Nexavar), a dual-actioninhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol. 2006; 407:597–612. [PubMed: 16757355]

103. Doerig C, Abdi A, Bland N, Eschenlauer S, Dorin-Semblat D, Fennell C, Halbert J, Holland Z,Nivez MP, Semblat JP, et al. Malaria: Targeting parasite and host cell kinomes. Biochim BiophysActa. 2010; 1804:604–612. [PubMed: 19840874]

104. Jirage D, Keenan SM, Waters NC. Exploring novel targets for antimalarial drug discovery:Plasmodial protein kinases. Infect Disord Drug Targets. 2010; 10:134–146. [PubMed: 20334624]

105. Leiriao P, Albuquerque SS, Corso S, van Gemert GJ, Sauerwein RW, Rodriguez A, Giordano S,Mota MM. HGF/MET signalling protects Plasmodium-infected host cells from apoptosis. CellMicrobiol. 2005; 7:603–609. [PubMed: 15760460]

106. Vangapandu S, Jain M, Kaur K, Patil P, Patel SR, Jain R. Recent advances in antimalarial drugdevelopment. Med Res Rev. 2007; 27:65–107. [PubMed: 16700012]

107. Khan MO, Levi MS, Tekwani BL, Khan SI, Kimura E, Borne RF. Synthesis and antimalarialactivities of cyclen 4-aminoquinoline analogs. Antimicrob Agents Chemother. 2009; 53:1320–1324. [PubMed: 19171802]

108. Sunduru N, Sharma M, Srivastava K, Rajakumar S, Puri SK, Saxena JK, Chauhan PM. Synthesisof oxalamide and triazine derivatives as a novel class of hybrid 4-aminoquinoline with potentantiplasmodial activity. Bioorg Med Chem. 2009; 17:6451–6462. [PubMed: 19665899]

109. Sunduru N, Srivastava K, Rajakumar S, Puri SK, Saxena JK, Chauhan PM. Synthesis of novelthiourea, thiazolidinedione and thioparabanic acid derivatives of 4-aminoquinoline as potentantimalarials. Bioorg Med Chem Lett. 2009; 19:2570–2573. [PubMed: 19339178]

110. Dow GS, Milner E, Caridha D, Gardner S, Lanteri C, Kozar M, Mannila A, McCalmont W,Melendez V, Moon J, et al. Central Nervous System (CNS) exposure of next generationquinoline methanols is reduced relative to mefloquine after intravenous (IV) dosing in mice. AmJ Trop Med Hyg. 2010; 83:213.

111. Milner E, McCalmont W, Bhonsle J, Caridha D, Carroll D, Gardner S, Gerena L, GettayacaminM, Lanteri C, Luong T, et al. Structure-activity relationships amongst 4-position quinolinemethanol antimalarials that inhibit the growth of drug sensitive and resistant strains ofPlasmodium falciparum. Bioorg Med Chem Lett. 2010; 20:1347–1351. [PubMed: 20097070]

112. Milner E, McCalmont W, Bhonsle J, Caridha D, Cobar J, Gardner S, Gerena L, Goodine D,Lanteri C, Melendez V, et al. Anti-malarial activity of a non-piperidine library of next-generationquinoline methanols. Malar J. 2010; 9:51. [PubMed: 20149249]

113. Tekwani BL, Walker LA. 8-Aminoquinolines: Future role as antiprotozoal drugs. Curr OpinInfect Dis. 2006; 19:623–631. [PubMed: 17075340]

114. Jefford CW. New developments in synthetic peroxidic drugs as artemisinin mimics. Drug DiscovToday. 2007; 12:487–495. [PubMed: 17532534]

115. Efferth T, Kaina B. Toxicity of the antimalarial artemisinin and its dervatives. Crit Rev Toxicol.2010; 40:405–421. [PubMed: 20158370]

116. Kongpatanakul S, Chatsiricharoenkul S, Khuhapinant A, Atipas S, Kaewkungwal J. Comparativestudy of dihydroartemisinin and artesunate safety in healthy Thai volunteers. Int J ClinPharmacol Ther. 2009; 47:579–586. [PubMed: 19761717]

117. Held J, Soomro SA, Kremsner PG, Jansen FH, Mordmuller B. In vitro activity of new artemisininderivatives against Plasmodium falciparum clinical isolates from Gabon. Int J AntimicrobAgents. 2011 submitted.

118. Sun L, Shah F, Helal MA, Wu Y, Pedduri Y, Chittiboyina AG, Gut J, Rosenthal PJ, Avery MA.Design, synthesis, and development of novel guaianolide-endoperoxides as potential antimalarialagents. J Med Chem. 2010; 53:7864–7868. [PubMed: 20945914]

Grimberg and Mehlotra Page 21

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 22: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

119. Posner GH, Chang W, Hess L, Woodard L, Sinishtaj S, Usera AR, Maio W, Rosenthal AS,Kalinda AS, D’Angelo JG, et al. Malaria-infected mice are cured by oral administration of newartemisinin derivatives. J Med Chem. 2008; 51:1035–1042. [PubMed: 18232653]

120. Singh AS, Verma VP, Hassam M, Krishna NN, Puri SK, Singh C. Amino- and hydroxy-functionalized 11-azaartemisinins and their derivatives. Org Lett. 2008; 10:5461–5464.[PubMed: 18991446]

121. Singh C, Chaudhary S, Kanchan R, Puri SK. Conversion of antimalarial drug artemisinin to a newseries of tricyclic 1,2,4-trioxanes1. Org Lett. 2007; 9:4327–4329. [PubMed: 17877363]

122. Singh C, Chaudhary S, Puri SK. New orally active derivatives of artemisinin with high efficacyagainst multidrug-resistant malaria in mice. J Med Chem. 2006; 49:7227–7233. [PubMed:17125275]

123. Dow GS, Koenig ML, Wolf L, Gerena L, Lopez-Sanchez M, Hudson TH, Bhattacharjee AK. Theantimalarial potential of 4-quinolinecarbinolamines may be limited due to neurotoxicity andcross-resistance in mefloquine-resistant Plasmodium falciparum strains. Antimicrob AgentsChemother. 2004; 48:2624–2632. [PubMed: 15215119]

124. Burgess SJ, Selzer A, Kelly JX, Smilkstein MJ, Riscoe MK, Peyton DH. A chloroquine-likemolecule designed to reverse resistance in Plasmodium falciparum. J Med Chem. 2006;49:5623–5625. [PubMed: 16942036]

125. Andrews S, Burgess SJ, Skaalrud D, Kelly JX, Peyton DH. Reversal agent and linker variants ofreversed chloroquines: Activities against Plasmodium falciparum. J Med Chem. 2010; 53:916–919. [PubMed: 20088608]

126. Burgess SJ, Kelly JX, Shomloo S, Wittlin S, Brun R, Liebmann K, Peyton DH. Synthesis,structure-activity relationship, and mode-of-action studies of antimalarial reversed chloroquinecompounds. J Med Chem. 2010; 53:6477–6489. [PubMed: 20684562]

127. Muregi FW, Ishih A. Next-generation antimalarial drugs: Hybrid molecules as a new strategy indrug design. Drug Dev Res. 2010; 71:20–32. [PubMed: 21399701]

128. Walsh JJ, Coughlan D, Heneghan N, Gaynor C, Bell A. A novel artemisinin-quinine hybrid withpotent antimalarial activity. Bioorg Med Chem Lett. 2007; 17:3599–3602. [PubMed: 17482816]

129. Cosledan F, Fraisse L, Pellet A, Guillou F, Mordmuller B, Kremsner PG, Moreno A, Mazier D,Maffrand JP, Meunier B. Selection of a trioxaquine as an antimalarial drug candidate. Proc NatlAcad Sci USA. 2008; 105:17579–17584. [PubMed: 18987321]

130. Newman DJ, Cragg GM, Snader KM. Natural products as sources of new drugs over the period1981–2002. J Nat Prod. 2003; 66:1022–1037. [PubMed: 12880330]

131. Ginsburg H, Deharo E. A call for using natural compounds in the development of newantimalarial treatments—An introduction. Malar J. 2011; 10(Suppl 1):S1. [PubMed: 21411010]

132. Fernandez LS, Sykes ML, Andrews KT, Avery VM. Antiparasitic activity of alkaloids from plantspecies of Papua New Guinea and Australia. Int J Antimicrob Agents. 2010; 36:275–279.[PubMed: 20580535]

133. Hnawia E, Hassani L, Deharo E, Maurel S, Waikedre J, Cabalion P, Bourdy G, Valentin A,Jullian V, Fogliani B. Antiplasmodial activity of New Caledonia and Vanuatu traditionalmedicines. Pharm Biol. 2011; 49:369–376. [PubMed: 21284535]

134. Kaur K, Jain M, Kaur T, Jain R. Antimalarials from nature. Bioorg Med Chem. 2009; 17:3229–3256. [PubMed: 19299148]

135. Turschner S, Efferth T. Drug resistance in Plasmodium: Natural products in the fight againstmalaria. Mini Rev Med Chem. 2009; 9:206–214. [PubMed: 19200025]

136. Wright CW. Recent developments in research on terrestrial plants used for the treatment ofmalaria. Nat Prod Rep. 2010; 27:961–968. [PubMed: 20532364]

137. Efferth T, Koch E. Complex interactions between phytochemicals. The multi-target therapeuticconcept of phytotherapy. Curr Drug Targets. 2011; 12:122–132. [PubMed: 20735354]

138. Bourdy G, Willcox ML, Ginsburg H, Rasoanaivo P, Graz B, Deharo E. Ethnopharmacology andmalaria: New hypothetical leads or old efficient antimalarials? Int J Parasitol. 2008; 38:33–41.[PubMed: 17720165]

Grimberg and Mehlotra Page 22

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 23: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

139. Willcox M, Benoit-Vical F, Fowler D, Bourdy G, Burford G, Giani S, Graziose R, Houghton P,Randrianarivelojosia M, Rasoanaivo P. Do ethnobotanical and laboratory data predict clinicalsafety and efficacy of anti-malarial plants? Malar J. 2011; 10(Suppl 1):S7. [PubMed: 21411018]

140. Wells TN. Natural products as starting points for future anti-malarial therapies: Going back to ourroots? Malar J. 2011; 10(Suppl 1):3. [PubMed: 21219614]

141. Plouffe D, Brinker A, McNamara C, Henson K, Kato N, Kuhen K, Nagle A, Adrian F, MatzenJT, Anderson P, et al. In silico activity profiling reveals the mechanism of action of antimalarialsdiscovered in a high-throughput screen. Proc Natl Acad Sci USA. 2008; 105:9059–9064.[PubMed: 18579783]

142. Gamo FJ, Sanz LM, Vidal J, de Cozar C, Alvarez E, Lavandera JL, Vanderwall DE, Green DV,Kumar V, Hasan S, et al. Thousands of chemical starting points for antimalarial leadidentification. Nature. 2010; 465:305–310. [PubMed: 20485427]

143. Guiguemde WA, Shelat AA, Bouck D, Duffy S, Crowther GJ, Davis PH, Smithson DC, ConnellyM, Clark J, Zhu F, et al. Chemical genetics of Plasmodium falciparum. Nature. 2010; 465:311–315. [PubMed: 20485428]

144. Rottmann M, McNamara C, Yeung BK, Lee MC, Zou B, Russell B, Seitz P, Plouffe DM, DhariaNV, Tan J, et al. Spiroindolones, a potent compound class for the treatment of malaria. Science.2010; 329:1175–1180. [PubMed: 20813948]

145. Rush MA, Baniecki ML, Mazitschek R, Cortese JF, Wiegand R, Clardy J, Wirth DF.Colorimetric high-throughput screen for detection of heme crystallization inhibitors. AntimicrobAgents Chemother. 2009; 53:2564–2568. [PubMed: 19307367]

146. Woynarowski JM, Krugliak M, Ginsburg H. Pharmacogenomic analyses of targeting the AT-richmalaria parasite genome with AT-specific alkylating drugs. Mol Biochem Parasitol. 2007;154:70–81. [PubMed: 17524501]

147. Fatumo S, Plaimas K, Mallm JP, Schramm G, Adebiyi E, Oswald M, Eils R, Konig R. Estimatingnovel potential drug targets of Plasmodium falciparum by analysing the metabolic network ofknock-out strains in silico. Infect Genet Evol. 2009; 9:351–358. [PubMed: 18313365]

148. Fatumo S, Plaimas K, Adebiyi E, Konig R. Comparing metabolic network models based ongenomic and automatically inferred enzyme information from Plasmodium and its human host todefine drug targets in silico. Infect Genet Evol. 2011; 11:201–208. [PubMed: 20804860]

149. Ekenna C, Fatuma S, Adebiyi E. In-silico evaluation of malaria drug targets. Int J EngineerTechnol. 2010; 2:132–135.

150. Wolf A, Shahid M, Kasam V, Ziegler W, Hofmann-Apitius M. In silico drug discoveryapproaches on grid computing infrastructures. Curr Clin Pharmacol. 2010; 5:37–46. [PubMed:19642967]

151. Degliesposti G, Kasam V, Da Costa A, Kang HK, Kim N, Kim DW, Breton V, Kim D, RastelliG. Design and discovery of plasmepsin II inhibitors using an automated workflow on large-scalegrids. ChemMedChem. 2009; 4:1164–1173. [PubMed: 19437467]

152. Kasam V, Zimmermann M, Maass A, Schwichtenberg H, Wolf A, Jacq N, Breton V, Hofmann-Apitius M. Design of new plasmepsin inhibitors: A virtual high throughput screening approachon the EGEE grid. J Chem Inf Model. 2007; 47:1818–1828. [PubMed: 17727268]

153. Kasam V, Salzemann J, Botha M, Dacosta A, Degliesposti G, Isea R, Kim D, Maass A, KenyonC, Rastelli G, Hofmann-Apitius M, et al. WISDOM-II: Screening against multiple targetsimplicated in malaria using computational grid infrastructures. Malar J. 2009; 8:88. [PubMed:19409081]

154. Gardner MJ, Hall N, Fung E, White O, Berriman M, Hyman RW, Carlton JM, Pain A, NelsonKE, Bowman S, et al. Genome sequence of the human malaria parasite Plasmodium falciparum.Nature. 2002; 419:498–511. [PubMed: 12368864]

155. Ntoumi F, Kwiatkowski DP, Diakite M, Mutabingwa TK, Duffy PE. New interventions formalaria: mining the human and parasite genomes. Am J Trop Med Hyg. 2007; 77(Suppl 6):70–275.

156. Ginsburg H. Progress in in silico functional genomics: The malaria Metabolic Pathways database.Trends Parasitol. 2006; 22:238–240. [PubMed: 16707276]

Grimberg and Mehlotra Page 23

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 24: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

157. Rao A, Yeleswarapu SJ, Raghavendra G, Srinivasan R, Bulusu G. PlasmoID: A P. falciparumprotein information discovery tool. In Silico Biol. 2009; 9:195–202. [PubMed: 20109149]

158. Ben Mamoun C, Prigge ST, Vial H. Targeting the lipid metabolic pathways for the treatment ofmalaria. Drug Dev Res. 2010; 71:44–55. [PubMed: 20559451]

159. Huthmacher C, Hoppe A, Bulik S, Holzhutter HG. Antimalarial drug targets in Plasmodiumfalciparum predicted by stage-specific metabolic network analysis. BMC Syst Biol. 2010; 4:120.[PubMed: 20807400]

160. Oyelade J, Ewejobi I, Brors B, Eils R, Adebiyi E. Computational identification of signallingpathways in Plasmodium falciparum. Infect Genet Evol. 201010.1016/j.meegid.2010.11.006

161. Mu J, Seydel KB, Bates A, Su XZ. Recent progress in functional genomic research inPlasmodium falciparum. Curr Genomics. 2010; 11:279–286. [PubMed: 21119892]

162. Choi SR, Mukherjee P, Avery MA. The fight against drug-resistant malaria: novel plasmodialtargets and antimalarial drugs. Curr Med Chem. 2008; 15:161–171. [PubMed: 18220771]

163. Dharia NV, Chatterjee A, Winzeler EA. Genomics and systems biology in malaria drugdiscovery. Curr Opin Investig Drugs. 2010; 11:131–138.

164. Gardiner DL, Skinner-Adams TS, Brown CL, Andrews KT, Stack CM, McCarthy JS, Dalton JP,Trenholme KR. Plasmodium falciparum: New molecular targets with potential for antimalarialdrug development. Expert Rev Anti-Infect Ther. 2009; 7:1087–1098. [PubMed: 19883329]

165. Jana S, Paliwal J. Novel molecular targets for antimalarial chemotherapy. Int J AntimicrobAgents. 2007; 30:4–10. [PubMed: 17339102]

166. Muregi FW, Kirira PG, Ishih A. Novel rational drug design strategies with potential torevolutionize malaria chemotherapy. Curr Med Chem. 2011; 18:113–143. [PubMed: 21110811]

167. Na-Bangchang K, Karbwang J. Current status of malaria chemotherapy and the role ofpharmacology in antimalarial drug research and development. Fundam Clin Pharmacol. 2009;23:387–409. [PubMed: 19709319]

168. Wells TN, Alonso PL, Gutteridge WE. New medicines to improve control and contribute to theeradication of malaria. Nat Rev Drug Discov. 2009; 8:879–891. [PubMed: 19834482]

169. Coteron JM, Catterick D, Castro J, Chaparro MJ, Diaz B, Fernandez E, Ferrer S, Gamo FJ, GordoM, Gut J, et al. Falcipain inhibitors: optimization studies of the 2-pyrimidinecarbonitrile leadseries. J Med Chem. 2010; 53:6129–6152. [PubMed: 20672841]

170. Gibbons P, Verissimo E, Araujo NC, Barton V, Nixon GL, Amewu RK, Chadwick J, Stocks PA,Biagini GA, Srivastava A, et al. Endoperoxide carbonyl falcipain 2/3 inhibitor hybrids: Towardcombination chemotherapy of malaria through a single chemical entity. J Med Chem. 2010;53:8202–8206. [PubMed: 20979352]

171. Shah F, Mukherjee P, Gut J, Legac J, Rosenthal PJ, Tekwani BL, Avery MA. Identification ofnovel malarial cysteine protease inhibitors using structure-based virtual screening of a focusedcysteine protease inhibitor library. J Chem Inf Model. 2011 submitted.

172. Teixeira C, Gomes JR, Gomes P. FalcipainsPlasmodium falciparum cysteine proteases as Keydrug targets against malaria. Curr Med Chem. 2011 submitted.

173. Pallavi R, Roy N, Nageshan RK, Talukdar P, Pavithra SR, Reddy R, Venketesh S, Kumar R,Gupta AK, Singh RK, et al. Heat shock protein 90 as a drug target against protozoan infections:biochemical characterization of HSP90 from Plasmodium falciparum and Trypanosoma evansiand evaluation of its inhibitor as a candidate drug. J Biol Chem. 2010; 285:37964–37975.[PubMed: 20837488]

174. Pesce ER, Cockburn IL, Goble JL, Stephens LL, Blatch GL. Malaria heat shock proteins: Drugtargets that chaperone other drug targets. Infect Disord Drug Targets. 2010; 10:147–157.[PubMed: 20334623]

175. Shonhai A. Plasmodial heat shock proteins: Targets for chemotherapy. FEMS Immunol MedMicrobiol. 2010; 58:61–74. [PubMed: 20041948]

176. Doerig C, Meijer L. Antimalarial drug discovery: Targeting protein kinases. Expert Opin TherTargets. 2007; 11:279–290. [PubMed: 17298288]

177. Doerig C, Billker O, Haystead T, Sharma P, Tobin AB, Waters NC. Protein kinases of malariaparasites: An update. Trends Parasitol. 2008; 24:570–577. [PubMed: 18845480]

Grimberg and Mehlotra Page 24

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 25: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

178. Holland Z, Prudent R, Reiser JB, Cochet C, Doerig C. Functional analysis of protein kinase CK2of the human malaria parasite Plasmodium falciparum. Eukaryot Cell. 2009; 8:388–397.[PubMed: 19114502]

179. Caridha D, Kathcart AK, Jirage D, Waters NC. Activity of substituted thiophene sulfonamidesagainst malarial and mammalian cyclin dependent protein kinases. Bioorg Med Chem Lett. 2010;20:3863–3867. [PubMed: 20627564]

180. Geyer JA, Keenan SM, Woodard CL, Thompson PA, Gerena L, Nichols DA, Gutteridge CE,Waters NC. Selective inhibition of Pfmrk, a Plasmodium falciparum CDK, by antimalarial 1,3-diaryl-2-propenones. Bioorg Med Chem Lett. 2009; 19:1982–1985. [PubMed: 19250824]

181. Geyer JA, Prigge ST, Waters NC. Targeting malaria with specific CDK inhibitors. BiochimBiophys Acta. 2005; 1754:160–170. [PubMed: 16185941]

182. Kato N, Sakata T, Breton G, Le Roch KG, Nagle A, Andersen C, Bursulaya B, Henson K,Johnson J, Kumar KA, et al. Gene expression signatures and small-molecule compounds link aprotein kinase to Plasmodium falciparum motility. Nat Chem Biol. 2008; 4:347–356. [PubMed:18454143]

183. Lemercier G, Fernandez-Montalvan A, Shaw JP, Kugelstadt D, Bomke J, Domostoj M, SchwarzMK, Scheer A, Kappes B, Leroy D. Identification and characterization of novel small moleculesas potent inhibitors of the plasmodial calcium-dependent protein kinase 1. Biochemistry. 2009;48:6379–6389. [PubMed: 19507895]

184. Bouloc N, Large JM, Smiljanic E, Whalley D, Ansell KH, Edlin CD, Bryans JS. Synthesis and invitro evaluation of imidazopyridazines as novel inhibitors of the malarial kinase PfPK7. BioorgMed Chem Lett. 2008; 18:5294–5298. [PubMed: 18774709]

185. Dorin-Semblat D, Sicard A, Doerig C, Ranford-Cartwright L, Doerig C. Disruption of the PfPK7gene impairs schizogony and sporogony in the human malaria parasite Plasmodium falciparum.Eukaryot Cell. 2008; 7:279–285. [PubMed: 18083830]

186. Klein M, Diner P, Dorin-Semblat D, Doerig C, Grotli M. Synthesis of 3-(1,2,3-triazol-1-yl)- and3-(1,2,3-triazol-4-yl)-substituted pyrazolo[3,4-d]pyrimidin-4-amines via click chemistry:potential inhibitors of the Plasmodium falciparum PfPK7 protein kinase. Org Biomol Chem.2009; 7:3421–3429. [PubMed: 19675896]

187. Merckx A, Echalier A, Langford K, Sicard A, Langsley G, Joore J, Doerig C, Noble M, EndicottJ. Structures of P. falciparum protein kinase 7 identify an activation motif and leads for inhibitordesign. Structure. 2008; 16:228–238. [PubMed: 18275814]

188. Dorin-Semblat D, Quashie N, Halbert J, Sicard A, Doerig C, Peat E, Ranford-Cartwright L,Doerig C. Functional characterization of both MAP kinases of the human malaria parasitePlasmodium falciparum by reverse genetics. Mol Microbiol. 2007; 65:1170–1180. [PubMed:17651389]

189. Reininger L, Tewari R, Fennell C, Holland Z, Goldring D, Ranford-Cartwright L, Billker O,Doerig C. An essential role for the Plasmodium Nek-2 Nima-related protein kinase in the sexualdevelopment of malaria parasites. J Biol Chem. 2009; 284:20858–20868. [PubMed: 19491095]

190. Wurtz N, Chapus C, Desplans J, Parzy D. cAMP-dependent protein kinase from Plasmodiumfalciparum: An update. Parasitology. 2011; 138:1–25. [PubMed: 20663247]

191. Andrews KT, Tran TN, Wheatley NC, Fairlie DP. Targeting histone deacetylase inhibitors foranti-malarial therapy. Curr Top Med Chem. 2009; 9:292–308. [PubMed: 19355992]

192. Rotili D, Simonetti G, Savarino A, Palamara AT, Migliaccio AR, Mai A. Non-cancer uses ofhistone deacetylase inhibitors: Effects on infectious diseases and beta-hemoglobinopathies. CurrTop Med Chem. 2009; 9:272–291. [PubMed: 19355991]

193. Chaal BK, Gupta AP, Wastuwidyaningtyas BD, Luah YH, Bozdech Z. Histone deacetylases playa major role in the transcriptional regulation of the Plasmodium falciparum life cycle. PLoSPathog. 2010; 6:e1000737. [PubMed: 20107518]

194. Andrews KT, Tran TN, Lucke AJ, Kahnberg P, Le GT, Boyle GM, Gardiner DL, Skinner-AdamsTS, Fairlie DP. Potent antimalarial activity of histone deacetylase inhibitor analogues.Antimicrob Agents Chemother. 2008; 52:1454–1461. [PubMed: 18212103]

Grimberg and Mehlotra Page 25

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 26: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

195. Wheatley NC, Andrews KT, Tran TL, Lucke AJ, Reid RC, Fairlie DP. Antimalarial histonedeacetylase inhibitors containing cinnamate or NSAID components. Bioorg Med Chem Lett.2010; 20:7080–7084. [PubMed: 20951583]

196. Dow GS, Chen Y, Andrews KT, Caridha D, Gerena L, Gettayacamin M, Johnson J, Li Q,Melendez V, Obaldia N 3rd, et al. Antimalarial activity of phenylthiazolyl-bearing hydroxamate-based histone deacetylase inhibitors. Antimicrob Agents Chemother. 2008; 52:3467–3477.[PubMed: 18644969]

197. Marfurt J, Chalfein F, Prayoga P, Wabiser F, Kenangalem E, Piera KA, Fairlie DP, Tjitra E,Anstey NM, Andrews KT, et al. Ex vivo activity of histone deacetylase inhibitors againstmultidrug-resistant clinical isolates of Plasmodium falciparum and P. vivax. Antimicrob AgentsChemother. 2011; 55:961–966. [PubMed: 21135175]

198. Painter HJ, Morrisey JM, Mather MW, Vaidya AB. Specific role of mitochondrial electrontransport in blood-stage Plasmodium falciparum. Nature. 2007; 446:88–91. [PubMed: 17330044]

199. Phillips MA, Rathod PK. Plasmodium dihydroorotate dehydrogenase: A promising target fornovel anti-malarial chemotherapy. Infect Disord Drug Targets. 2010; 10:226–239. [PubMed:20334617]

200. Rodrigues T, Lopes F, Moreira R. Inhibitors of the mitochondrial electron transport chain and denovo pyrimidine biosynthesis as antimalarials: The present status. Curr Med Chem. 2010;17:929–956. [PubMed: 20156168]

201. Booker ML, Bastos CM, Kramer ML, Barker RH Jr, Skerlj R, Sidhu AB, Deng X, Celatka C,Cortese JF, Guerrero Bravo JE, et al. Novel inhibitors of Plasmodium falciparum dihydroorotatedehydrogenase with anti-malarial activity in the mouse model. J Biol Chem. 2010; 285:33054–33064. [PubMed: 20702404]

202. Davies M, Heikkila T, McConkey GA, Fishwick CW, Parsons MR, Johnson AP. Structure-baseddesign, synthesis, and characterization of inhibitors of human and Plasmodium falciparumdihydroorotate dehydrogenases. J Med Chem. 2009; 52:2683–2693. [PubMed: 19351152]

203. Heikkila T, Ramsey C, Davies M, Galtier C, Stead AM, Johnson AP, Fishwick CW, Boa AN,McConkey GA. Design and synthesis of potent inhibitors of the malaria parasite dihydroorotatedehydrogenase. J Med Chem. 2007; 50:186–191. [PubMed: 17228860]

204. Ojha PK, Roy K. Chemometric modeling, docking and in silico design of triazolopyrimidine-based dihydroorotate dehydrogenase inhibitors as antimalarials. Eur J Med Chem. 2010;45:4645–4656. [PubMed: 20708304]

205. Patel V, Booker M, Kramer M, Ross L, Celatka CA, Kennedy LM, Dvorin JD, Duraisingh MT,Sliz P, Wirth DF, et al. Identification and characterization of small molecule inhibitors ofPlasmodium falciparum dihydroorotate dehydrogenase. J Biol Chem. 2008; 283:35078–35085.[PubMed: 18842591]

206. Rowe JA, Claessens A, Corrigan RA, Arman M. Adhesion of Plasmodium falciparum-infectederythrocytes to human cells: molecular mechanisms and therapeutic implications. Expert RevMol Med. 2009; 11:e16. [PubMed: 19467172]

207. Nathoo S, Serghides L, Kain KC. Effect of HIV-1 antiretroviral drugs on cytoadherence andphagocytic clearance of Plasmodium falciparum-parasitised erythrocytes. Lancet. 2003;362:1039–1041. [PubMed: 14522535]

208. Adams Y, Smith SL, Schwartz-Albiez R, Andrews KT. Carrageenans inhibit the in vitro growthof Plasmodium falciparum and cytoadhesion to CD36. Parasitol Res. 2005; 97:290–294.[PubMed: 16012863]

209. Dondorp AM, Silamut K, Charunwatthana P, Chuasuwanchai S, Ruangveerayut R, Krintratun S,White NJ, Ho M, Day NP. Levamisole inhibits sequestration of infected red blood cells inpatients with falciparum malaria. J Infect Dis. 2007; 196:460–466. [PubMed: 17597461]

210. Dormeyer M, Adams Y, Kramer B, Chakravorty S, Tse MT, Pegoraro S, Whittaker L, Lanzer M,Craig A. Rational design of anticytoadherence inhibitors for Plasmodium falciparum based onthe crystal structure of human intercellular adhesion molecule 1. Antimicrob Agents Chemother.2006; 50:724–730. [PubMed: 16436732]

Grimberg and Mehlotra Page 26

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 27: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

211. Andrews KT, Klatt N, Adams Y, Mischnick P, Schwartz-Albiez R. Inhibition of chondroitin-4-sulfate-specific adhesion of Plasmodium falciparum-infected erythrocytes by sulfatedpolysaccharides. Infect Immun. 2005; 73:4288–4294. [PubMed: 15972521]

212. Simmons DL. Anti-adhesion therapies. Curr Opin Pharmacol. 2005; 5:398–404. [PubMed:15955735]

213. Kerb R, Fux R, Morike K, Kremsner PG, Gil JP, Gleiter CH, Schwab M. Pharmacogenetics ofantimalarial drugs: Effect on metabolism and transport. Lancet Infect Dis. 2009; 9:760–774.[PubMed: 19926036]

Grimberg and Mehlotra Page 27

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 28: Author Manuscript NIH Public Access Rajeev K. Mehlotra ...aquadoc.typepad.com/files/grimberg-antimalarial-rx-2011.pdfMore than 40% of the world’s population, much of it socioeconomically

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Grimberg and Mehlotra Page 28

Tabl

e 1

Rec

ent a

ntim

alar

ial d

rug

scre

enin

g ef

forts

.

Gro

upN

umbe

r of

com

poun

ds sc

reen

edN

umbe

r of

hits

(effe

ctiv

e ag

ains

t mal

aria

para

site

s)N

umbe

r of

new

lead

s (w

ith n

o re

port

ed to

xici

ty to

hum

ans)

Pre-

IND

Ref

eren

ce

Plou

ffe

et a

l.1,

700,

000

6,00

053

0*

[141

]

Gam

o et

al.

2,00

0,00

013

,533

51*

[142

]

Gui

guem

de e

t al.

309,

474

1,30

017

234

[143

]

Rot

tman

n et

al.

12,0

0027

517

1[1

44]

Rus

h et

al.

16,0

0017

**

[145

]

Grim

berg

et a

l.33

286

3[8

8]

* Ong

oing

inve

stig

atio

ns, o

utco

me

pend

ing.

Pharmaceuticals (Basel). Author manuscript; available in PMC 2011 May 26.