656
Views
169
CrossRef citations to date
0
Altmetric
Review

Drugs in development for toxoplasmosis: advances, challenges, and current status

&
Pages 273-293 | Published online: 25 Jan 2017

Abstract

Toxoplasma gondii causes fatal and debilitating brain and eye diseases. Medicines that are currently used to treat toxoplasmosis commonly have toxic side effects and require prolonged courses that range from weeks to more than a year. The need for long treatment durations and the risk of relapsing disease are in part due to the lack of efficacy against T. gondii tissue cysts. The challenges for developing a more effective treatment for toxoplasmosis include decreasing toxicity, achieving therapeutic concentrations in the brain and eye, shortening duration, eliminating tissue cysts from the host, safety in pregnancy, and creating a formulation that is inexpensive and practical for use in resource-poor areas of the world. Over the last decade, significant progress has been made in identifying and developing new compounds for the treatment of toxoplasmosis. Unlike clinically used medicines that were repurposed for toxoplasmosis, these compounds have been optimized for efficacy against toxoplasmosis during preclinical development. Medicines with enhanced efficacy as well as features that address the unique aspects of toxoplasmosis have the potential to greatly improve toxoplasmosis therapy. This review discusses the facets of toxoplasmosis that are pertinent to drug design and the advances, challenges, and current status of preclinical drug research for toxoplasmosis.

Toxoplasma gondii

Toxoplasma gondii is a protozoan parasite that belongs to the phylum Apicomplexa. Apicomplexa also includes the medically important genera, Plasmodium, Babesia, and Cryptosporidium. The eponymous organelle of Apicomplexa, the apical complex, is used to invade the host cell. Biological similarities among apicomplexans are the basis for shared susceptibility to drugs such as the antifolate drugs pyrimethamine and sulfonamides and the anti-respiratory drug atovaquone. The current treatments for T. gondii and Babesia microti are drugs that were used as anti-malarials prior to being repurposed. However, an examination of parasite genomes, routes of infection, life cycle stages, hosts, and disease manifestations reveals diversity in the underlying biology of apicomplexan pathogens. Drugs that are specifically designed to optimize the efficacy against T. gondii hold potential for improving the treatment of toxoplasmosis.

The unique pathogenesis of T. gondii also presents challenges for drug therapy. Unlike many apicomplexans, T. gondii crosses the blood–brain barrier and establishes persistent infection in a drug-resistant bradyzoite stage. An ideal medicine for toxoplasmosis would achieve therapeutic, systemic, brain and eye concentrations to be effective in the organs where the majority of disease occurs and would be active against both the acute replicating tachyzoite and latent bradyzoite stages of the parasite. New drugs should also prioritize having fewer, milder side effects, a significant problem with the current first-line drugs. Current research into new drugs developed specifically for toxoplasmosis has led to promising preclinical compounds. This review discusses the aspects of toxoplasmosis that are germane to drug development and ongoing preclinical drug research.

T. gondii is a remarkably successful parasite that is broadly distributed throughout the world and is capable of infecting both mammals and birds. Up to one-third of the human population is estimated to have been infected.Citation1 The great majority of human T. gondii infection occurs either by ingestion of oocysts that are generated in the felid intestine and spread throughout the environment via feces or ingestion of T. gondii tissue cysts in undercooked meat. Congenital infection occurs through vertical transmission when a previously uninfected mother is infected during pregnancy. Otherwise, uncommon means of transmission include transplantation of infected organs, blood transfusion, or inhalation of oocyst-contaminated dust.Citation2 The resiliency of T. gondii oocysts in the environment contributes to the high rates of T. gondii infection in humans, and the risk of toxoplasmosis outbreaks, as evidenced by large waterborne outbreaks of T. gondii infection from oocyst-contaminated drinking water in Canada and Brazil.Citation3,Citation4 Although preventive measures focused on hygiene and sanitary meat production may have reduced the prevalence of human T. gondii infection, these measures will not reduce the overall burden of human T. gondii infection enough to decrease the need for better anti-Toxoplasma therapies in the near future.

Prevalence of T. gondii infection and disease

The seroprevalence of T. gondii antibodies varies significantly worldwide as rates of human infection are influenced by climate, the consumption of undercooked meat, hygiene, and exposure to cats.Citation5 Direct comparisons of seroprevalence studies are limited by heterogeneous methodologies, but have been important in identifying specific high prevalence populations. For example, seropositivity for T. gondii antibodies in Brazil ranges from 20% to >90% among different groups.Citation6 In the US, the seroprevalence among people aged 12–49 years has declined from 14.1% to 6.7% between 1994 and 2010.Citation7 However, prevalence in the US was reported to be 29.9% in people aged >70 years and 25.1% in US residents born outside of the US.Citation7 In studies of pregnant women and women of child bearing age, seroprevalence in Europe, Asia, and Africa ranges from 20% to 60%.Citation5

Although studies of seroprevalence provide valuable insight into T. gondii transmission and the underlying risk for the development of toxoplasmosis in a population, the worldwide incidence of disease caused by T. gondii is less understood. In addition to the risk of acquisition of T. gondii infection, the development of symptomatic toxoplasmosis is influenced by the prevalence of immunosuppressive conditions such as AIDS and may be influenced by differences in the virulence of T. gondii strains found all over the world. The hypothesis that certain strains of T. gondii are more virulent is most evident in Brazil where non-archetypal strains are associated with symptomatic ocular toxoplasmosisCitation8 and in French Guiana with severe disseminated disease in the immunocompetent.Citation9 By comparison, 1%–2% of persons infected with T. gondii develop eye disease in the US, whereas eye disease can approach up to 20% of seropositive persons in highly endemic areas of Brazil.Citation10 The broad distribution of T. gondii disease in populations with limited medical resources indicates that a new anti-T. gondii drug should be orally bioavailable, have a chemically stable formulation, and be inexpensive to produce.

Toxoplasmosis

T. gondii infection in immunocompetent hosts rarely requires drug therapy. Over 80% of primary T. gondii infections in immunocompetent hosts are asymptomatic.Citation11 Symptomatic infection typically consists of self-limited bilateral nontender cervical lymphadenopathy, which may be accompanied by fevers and myalgias. Following primary infection, T. gondii establishes latent infection, converting to the quiescent bradyzoite form within tissue cysts. Although the host immune system is capable of controlling most T. gondii primary infections, ocular cysts may be reactivated and lead to vision loss from retinal scarring. In the US, 21,000 persons per year are estimated to develop ocular lesions and 4,800 persons per year develop symptomatic ocular lesions.Citation10 The effect of latent T. gondii brain infection on human behavior and mental health in immunocompetent individuals is an active area of research, but a causal relationship between latent infection and psychiatric disease or changes in human behavior has not been established.Citation12Citation14 Unlike T. gondii in most healthy people, infection in the immunocompromised is often debilitating or fatal.

Severe manifestations of T. gondii most often occur when tissue cysts reactivate in the setting of immunosuppression due to AIDS or medical immunosuppression. The most frequent severe clinical presentation is Toxoplasma encephalitis, which typically consists of multiple discrete brain lesions. Ocular and pulmonary diseases are the most common extra-cerebral sites of infection; however, disease involving other organs has been reported as well as sepsis due to disseminated disease.Citation15 In individuals with AIDS and a CD4 T-cell count <100 cells/µL, the incidence of encephalitis is 28% in seropositive patients not taking prophylaxis.Citation16 In allogeneic stem cell transplant patients, the 6-month incidence of symptomatic toxoplasmosis has been reported to be as high as 6% in a prospective multicenter study of seropositive patients, although prior retrospective studies have reported a lower incidence.Citation17Citation20 Of note, patients who developed symptomatic toxoplasmosis in this study were not taking standard prophylaxis. Toxoplasmosis has been reported in autologous stem cell transplant patients as well, although the risk for disease is lower.Citation21 Despite the relatively low prevalence of T. gondii infection in the US, in 2008, there were ~3,585 toxoplasmosis-related hospitalizations.Citation22 The cost of illness in the US caused by T. gondii has been estimated to be ~$3 billion and an annual loss of 11,000 quality-adjusted life years.Citation23 Severe toxoplasmosis in the US is reduced from the height of the AIDS epidemic prior to highly active antiretroviral therapy, but remains a significant source of morbidity and mortality in the US and worldwide.

Severe toxoplasmosis also results from congenital infection and may occur rarely in healthy individuals. Congenital toxoplasmosis ranges from subclinical to severe disease that consists primarily of brain and eye manifestations, but may also include extracranial pathology in up to half of neonates.Citation24 Although rare, severe toxoplasmosis in the immunocompetent has been reported, and in a notable cluster of cases in French Guiana, it seems to be due to virulent, non-archetypal strains.Citation9,Citation25Citation27 Unlike typical toxoplasmosis, patients in these series presented with disseminated infection involving multiple organs, frequently had pulmonary involvement, and required intensive care.

Current medicines for toxoplasmosis

First-line therapy consists of the combination of pyrimethamine and sulfadiazine with leucovorin added to prevent hematologic toxicity. In observational studies and controlled trials for Toxoplasma encephalitis, this regimen has been found to have high rates of toxic side effects leading to discontinuation of therapy. A review of 115 patients with Toxoplasma encephalitis found toxicity in 62% of patients and severe side effects requiring a change in therapy in 44% of patients.Citation28 Similarly, in treatment trials of Toxoplasma encephalitis, toxicity led to discontinuation of pyrimethamine-sulfadiazine in one-third of patients.Citation29,Citation30 Sulfadiazine may be replaced with clindamycin if the patient has an allergy to sulfa drugs; however, the clindamycin-containing regimen is less effective in preventing relapse and had similar rates of toxicity.Citation29 Trimethoprim-sulfamethoxazole has been shown to have efficacy similar to pyrimethamine-sulfadiazine and may be used as an alternative if patients do not have sulfa allergy and pyrimethamine is not tolerated or is not available.Citation31Citation33 Atovaquone or azithromycin may be used as alternate therapy in combination with pyrimethamine or sulfadiazine for the treatment and prophylaxis of toxoplasmosis when first-line therapy is contraindicated. However, use of these alternate therapies is supported by less clinical data, and these regimens have similar rates of patient intolerance.Citation34Citation36 In addition to frequent side effects, pyrimethamine and sulfadiazine are associated with rare severe reactions that may be fatal, including agranulocytosis, Stevens–Johnson syndrome, toxic epidermal necrolysis, and hepatic necrosis. Drugs that are less toxic would greatly improve the care of patients with toxoplasmosis.

The need for nontoxic medicines is further emphasized by the prolonged courses of therapy required for treatment and suppression of infection. Initial treatment duration for Toxoplasma encephalitis is at least 6 weeks followed by secondary suppression until sufficient immune reconstitution; duration for congenital infection is at least 1 year; ocular infection is 4–6 weeks with the consideration of continued suppression to prevent relapse.Citation24,Citation37,Citation38 Prolonged courses are required, in part because current clinical medicines are unable to eliminate the tissue-cyst stage of T. gondii. A promising aspect of several experimental compounds is activity against T. gondii tissue cysts that are established in mice 5 weeks prior to treatment.Citation39,Citation40 Previous studies have shown that atovaquone possesses activity in experimental models in vivo against tissue cysts; however, clinical studies of ocular infection and encephalitis have not shown an advantage in preventing relapse, which may be related to subtherapeutic drug concentrations in infected tissue compartments or translational limitations of toxoplasmosis animal models.Citation34,Citation38,Citation41

Challenges for anti-T. gondii drugs

The biology of T. gondii bradyzoite containing tissue cysts has been poorly understood and is an active area of research that will be important for the development of drugs that eliminate T. gondii infection from the host. Tissue cysts are made up of bradyzoites surrounded by a thick glycan-rich cyst wall. Animal models have provided important insights into drugs that have the potential to cure latent T. gondii infection. However, virulence and cyst formation vary depending on the T. gondii strain and the host species and strain. It is unclear which animal model most resembles human infection. In addition to uncertainty regarding the most appropriate translational animal models, the key biological questions of the metabolic pathways that are essential for bradyzoite survival in the tissue cyst and which of these are viable drug targets remain mostly unanswered. McPhillie et alCitation42 made recent progress in understanding bradyzoite biology, demonstrating that the cyst-forming EGS strain possesses a mutation in the transcription factor Apetala 2, a known repressor of bradyzoite genes. Provocative studies of the ME49 T. gondii strain in CBA/J mice have shown heterogeneous levels of replication among bradyzoites within cysts and variation between cysts.Citation43 Future studies probing the biology and drug susceptibility of latent T. gondii infection should provide more effective strategies for eliminating T. gondii tissue cysts from the host, preventing relapsing disease and potentially shortening the required duration of drug administration.

Drug resistance in T. gondii is suspected to contribute to treatment failures that have occurred in ~10% of patients during initial therapy and 10%–20% of patients who relapse during suppressive therapy. However, the contribution of drug resistance to treatment failure is difficult to quantify because T. gondii is not routinely recovered from patients with toxoplasmosis. Moreover, treatment failures of toxoplasmosis during clinical studies have been difficult to characterize due to diagnostic limitations, drug intolerance, nonadherence, variable drug absorption, and the complex morbidities of patients.Citation29,Citation30,Citation32,Citation34,Citation44 High-level sulfonamide resistance has been found in clinical isolates without known sulfonamide exposure and after sulfadoxine exposure.Citation45,Citation46 The resistant T. gondii strain in which there was no drug exposure possessed a mutation in the dihydropteroate synthase (DHPS) gene known to confer resistance in both T. gondii and Plasmodium falciparum and demonstrated cross-resistance to several sulfonamides including sulfadiazine and sulfamethoxazole.Citation43 However, genetic mutations in DHPS do not account for sulfonamide resistance in all clinical isolates, and research into alternate mechanisms is ongoing.Citation45,Citation47,Citation48

The range of sulfonamide susceptibility seems to be greater than that of pyrimethamine or atovaquone. Genetic evidence of atovaquone resistance has not been found in clinical isolates, but has been obtained in vitro with chemical mutagenesis.Citation49 Variations in atovaquone susceptibility among different T. gondii genotypes have been observed but subsequent experiments did not find significant differences in genotype susceptibility.Citation45,Citation50 Similarly, type I T. gondii strains have been shown to be less susceptible to pyrimethamine in vitro compared to type II and type III strains; however, subsequent studies found that decreased susceptibility to pyrimethamine correlated with the replication rate rather than genotype and decreased susceptibility was not great enough to be considered significant in pyrimethamine resistance.Citation45,Citation51 Differences in in vitro susceptibility for pyrimethamine and atovaquone found in these studies likely reflect the differences in methodologies, and the translational significance of these findings is not clear. However, given that sulfonamides are part of first-line therapy, variations in sulfonamide susceptibility, whether preexisting or occurring after drug exposure are concerning and further accentuate the need for new drugs for toxoplasmosis.

Preclinical testing of anti-toxoplasma compounds

Well-established in vitro assays and animal models exist for preclinical anti-T. gondii drug development, but there is no standard testing cascade. Most often, anti-Toxoplasma compounds are initially evaluated in vitro in tachyzoite replication assays. Common assays use T. gondii that expresses β-galactosidase or fluorescent proteins, or plaque formation or [3H] uracil uptake in non-transgenic T. gondii. T. gondii requires a host cell to replicate; therefore, it is also important to assess the effects of the compounds on the viability of the host cells while assessing the degree of compound inhibition. Although T. gondii inhibition assays may be performed on different strains of T. gondii, rates of replication vary among different strains, and the type I RH strain is often used initially because it is well adapted to in vitro culture and replicates relatively quickly and reliably.

The RH strain is typically fatal in mice within 10 days providing a model that reflects fulminate, disseminated infection that is not controlled by the host immune system. This strain is useful to evaluate acute infection; however, this model does not evaluate activity against brain tissue cysts, and type I strains are not the dominant strain in most of the areas of the world. Type II strains are more prominent in North America and Europe, are less virulent in mice, and establish tissue cysts in the mouse brain. Susceptibility to infection with type II strains differs among mouse strains, and the degree to which tissue cysts in the mouse brain resemble latent human T. gondii infection is unknown. Recent experiments have indicated that the ongoing replication of bradyzoites occurs within the established brain tissue cysts as long as 8 weeks after infection drawing into question the presumption that latent bradyzoite infection is metabolically dormant.Citation43,Citation52 Models of latent infection that started treatment with experimental compounds after 5 weeks of sublethal infection with the ME49 strain have demonstrated brain cyst reduction by experimental compounds.Citation39,Citation53 In addition to acute and latent murine models of infection, congenital and ocular toxoplasmosis models as well as other animal species have been evaluated, but the majority of experimental compounds are initially tested in models wherein mice are infected orally or by intraperitoneal injection, and parasite burden in tissues or survival is measured as outcome.Citation54

Future testing of preclinical compounds against genetically diverse, non-archetypal strains of T. gondii both in vitro and in vivo should prove insightful in that virulence and pathogenesis vary between strains. Recently characterized parasite strains, such as the EGS strain that forms thick-walled cysts spontaneously in tissue culture allows for the study of inhibitors against cysts in vitro.Citation42 In addition, this strain has been genetically modified to express stage-specific fluorescence to further characterize the activity against bradyzoites and tissue cysts.Citation55 Beyond improvements in testing cascades, new methods of target identification are likely to contribute to drug discovery. Future investigation of drug targets in Toxoplasma has recently been advanced by a genome-wide screen using CRISPR/Cas9 to identify essential genes.Citation56 Using the phenotypic data from this screen, which is available through www.toxodb.org, researchers are able to investigate fitness-conferring proteins for potential drug targets.Citation57

Basic research in T. gondii biology and drug screening has identified a diverse array of drug targets. Over 20 preclinical drug development projects have been described in publications over the past decade. The research studies selected in this review reflect the advances, challenges, and current status of drug development for toxoplasmosis. presents the classes of compounds in alphabetical order as they appear in the text and with the chemical structures.

Table 1 Experimental compounds for toxoplasmosis

Artemisinin derivatives

Derivatives of artemisinin, an endoperoxidase, are the backbone of first-line treatment for malaria. Artemisinin and its derivatives also have in vitro efficacy against T. gondii.Citation58Citation60 Anti-parasitic activity of these compounds requires a 1,2,4-trioxane moiety and is enhanced by substitutions at the C-10 position.Citation61

Schultz et alCitation62 tested two artemisinin derivatives, CPH4–136 and LEW3–27, in murine models of acute and chronic infection. CPH4–136 and LEW3–27 contain thiazole and carboxamide substitutions at their respective C-10 positions and were previously found to have in vitro activity against tachyzoites (340 and 360 nM, respectively).Citation63 Mice were infected intraperitoneally (IP) with RH strain tachyzoites and then treated with 3–50 mg/kg of either compound IP for 7 days. Both the compounds prolonged survival to a modest extent. Approximately 20% of infected mice treated with LEW3–27 at 30 mg/kg/day were alive at day 20, and all the mice treated with CPH4–136 at 10 mg/kg/day were dead by day 18. All the infected mice treated with atovaquone (10 mg/kg/day) survived until day 20, and the infected, untreated mice died by 12 days post-infection. In addition, CPH4–136 at doses of 30 and 10 mg/kg was associated with early mortality in the treatment group compared to the control group treated with vehicle alone, raising questions regarding toxicity at these doses. However, in a murine model of infection with the ME49 strain, a lower 3 mg/kg dose of CPH4–136 reduced cyst burden by ~40% when given for 32 days. Neither LEW3–27 nor artemether reduced the cyst burden.

Bisphosphonates: farnysyl diphosphate inhibitors

Bisphosphonates are used for the treatment of osteoporosis and other diseases of bone resorption. Nitrogen-containing bisphosphonates, such as, alendronate or risedronate, inhibit farnesyl pyrophosphate synthase in the mevalonate pathway in osteoclasts.Citation64 Many parasitic protozoa, including T. gondii, possess a mevalonate pathway for the synthesis of sterols and polyisoprenoids. A key branch of this pathway is the synthesis of farnesyl diphosphate, a precursor to ubiquinone, sterols, and prenylated proteins. Synthesis of farnesyl diphosphate in T. gondii is catalyzed by the bifunctional enzyme farnesyl diphosphate/geranylgeranyl-diphosphate synthaseCitation65 and is inhibited by nitrogen-containing bisphosphonates.Citation66

In an early study,Citation66 risedronate was shown to have an in vitro IC50 of 490 nM against T. gondii. In vivo experimentsCitation67 demonstrated that risedronate (10 mg/kg/day) improved 30-day survival by 35% in Swiss Webster mice infected orally with 10 cysts of the C56 strain of T. gondii. An amount of 20 mg/kg/day improved 30-day survival by 55%. Later workCitation67 further improved the potency of bisphosphonate derivatives. Compound 1 in the series described in Ling et al is an n-alkyl derivative that has an in vitro IC50 of 280 nM. When administered to mice infected with 5 cysts of C56 T. gondii at 10 mg/kg IP daily for 10 days, compound 1 improved 30-day survival to 80%.

Bumped kinase inhibitors (BKIs)

BKIs inhibit the T. gondii calcium-dependent protein kinase 1 (TgCDPK1), a member of the serine/threonine protein kinase family.Citation68,Citation69 TgCDPK1 regulates the calcium-dependent pathway of T. gondii microneme secretion and is required for gliding motility, host-cell invasion, and egress.Citation70 BKIs inhibit T. gondii replication by blocking host-cell invasion and egress.Citation71,Citation72 T. gondii mitogen-activated protein kinase like 1 (TgMAPKL1) has also been suggested as a secondary target for the BKI, 1NM-PP1, as a mutation in TgMAPKL1, was associated with decreased susceptibility to 1NM-PP1 and similar BKIs.Citation73,Citation74

A key structural difference between TgCDPK1 and human kinases occurs at the “gatekeeper residue” in the ATP-binding pocket. TgCDPK1 contains a small glycine residue at this position, whereas human kinases have larger residues. The additional space afforded by the glycine residue in TgCDPK1 has been exploited for the design of potent and selective ATP-competitive TgCDPK1 inhibitors.Citation68,Citation75 Numerous BKI analogs have been developed around different core scaffolds that have promising in vitro and in vivo activities.Citation53,Citation76Citation79

Potent TgCDPK1 inhibitors have been synthesized from pyrazolopyrimidine (PP) and 5-aminopyrazole-4-carboxamide (AC) scaffolds.Citation53,Citation71,Citation72,Citation74,Citation79,Citation80Citation82 The BKI scaffold binds in the ATP-binding pocket of TgCDPK1 and the addition of a 6-alkoxy-2-naphthyl group at the C-3 position, and a 4-piperidinylmethylene group at the N-1 position of the PP scaffold further increases selectivity.Citation75,Citation76 These BKIs were >15,000-fold more active against TgCDPK1 compared to the human kinases SRC and ABL, with no inhibition at 20 µM. BKI 1294, which was synthesized with N-methylation of the 4-piperidinyl-methylene substituent to improve metabolic stability, had an in vitro IC50 of 140 nM and reduced acute T. gondii infection by 93% when given orally at 30 mg/kg.Citation76 Later, BKI 1294 was found to inhibit the human Ether-à-go-go-Related Gene (hERG) ion channel preventing further advancement due to the risk of cardiotoxicity. Vidadala et al investigated the modifications of PP scaffold that maintained efficacy and selectivity for TgCDPK1 while eliminating hERG liability.Citation53 Compound 32 in their series of BKIs with reduced hERG activity had a >10 µM IC50 against hERG, a 60 nM in vitro IC50 against T. gondii and was effective in vivo. Compound 32 administered orally to mice at 20 mg/kg for 5 days eliminated acute RH strain peritoneal infection. In the latent T. gondii infection model using the ME49 strain, 30 mg/kg of compound 32 given orally reduced the number of brain cysts by 88.7%. Pharmacokinetic studies in noninfected mice demonstrated a brain to plasma concentration ratio of 0.33.Citation53 Lourido et alCitation77 described a series of PP analogs. Compounds 11 and 24 were metabolically stable and had in vitro IC50s of 250 and 610 nM. These compounds increased 30-day survival in mice infected with type II Pru-LUC strain and decreased brain cysts when administered IP prior to infection at 5 mg/kg for 10 days.Citation77

Development of the AC scaffold by Huang et al identified compounds 34 and 35 as lead compounds. Compound 35 had an in vitro IC50 of 89 nM against T. gondii, and in an acute in vivo model using the RH strain, compound 35 reduced infection in the peritoneum below the limits of detection when given orally at 20 mg/kg.Citation79 Compounds 34 and 35 had a brain to plasma concentration ratio of 0.16 and 0.43, respectively.

In addition to the PP and AC scaffolds, Moine et al identified biphenylimidazoazines as T. gondii growth inhibitors by screening a library of compounds.Citation82 Later, a series of 14 imidazo[1,2-b]pyridazines based on the biphenylimidazoazines were found to inhibit TgCDPK1 in enzymatic studies at EC50s of <1 µM and 7 compounds inhibited T. gondii in vitro at IC50s of <1 µM. Compound 1, 16a and 16f demonstrated in vitro IC50s of 50 nM, 100 nM and 70 nM, respectively, with minimal host cell toxicity.Citation83

Dihydrofolate reductase (DHFR) inhibitors

T. gondii, like several other protozoal parasites, has a unique bifunctional DHFR–thymidylate synthase (TS) that contains both catalytic sites on the same protein. DHFR is an extensively studied drug target that is inhibited by pyrimethamine, one of the first-line agents currently used against toxoplasmosis. Clinical use of pyrimethamine is primarily limited by the inhibition of host folate metabolism, which may cause neutropenia. DHFR inhibitors that have greater selectivity for T. gondiiCitation84 could potentially be less toxic and more effective.

The dihydrotriazines are a new class of DHFR inhibitors originally developed for use against malariaCitation85,Citation86 that have also been found to be effective against T. gondii. The dihydrotriazine JPC-2067-BCitation87 inhibited in vitro growth of T. gondii with an IC50 of 20 nM. JPC-2067-B has poor oral bioavailability but was effective in a murine model of acute toxoplasmosis when given IP at a dose of 1.25 mg/kg/day for 4 days. Likewise, the orally available prodrug of JPC-2067-B, JPC-2056, was effective in the same murine model at an oral dose of 40 mg/kg twice daily for 3 days.

Endoplasmic reticulum-associated degradation (ERAD) inhibitors

Proteins that are inserted into a membrane or secreted from the cell undergo folding and post-translational modification in the ER. A small fraction of proteins become irreversibly misfolded, and these proteins are recycled by a ubiquitin- and proteasome-mediated process called ERAD.Citation88 Most eukaryotic cells possess an extensive system for detecting misfolded proteins and target them to the ERAD pathway. However, the ERAD system in T. gondii, P. falciparum, and other protozoan parasites is limited relative to other eukaryotesCitation89 rendering these parasites more susceptible to interference with this pathway. Harbut et alCitation89 screened inhibitors known to target various proteins in the ERAD pathway for activity against P. falciparum. The authors determined that inhibitors of signal peptide peptidase (SPP) were the most effective and nontoxic agents based on in vitro IC50s against P. falciparum and human hepatocytes. One of these compounds, NITD731, inhibited T. gondii in human U-2 osteosarcoma (OS) cell culture at an IC50 of 71 nM. No toxicity to host cells was noted at a compound concentration of 10 µM. These results demonstrate that SPP is a potential target for apicomplexan inhibitors.

Fatty acid synthesis inhibitors

Synthesis of fatty acids in T. gondii takes place in the apicoplast.Citation90Citation92 Consistent with the evolutionary origin of this organelle, iterative elongation of nascent fatty acids is catalyzed by the multienzyme fatty acid synthase II (FAS II) pathway also found in bacteria and plants.Citation91,Citation93,Citation94 This is in contrast to the FAS I pathway found in animals and fungi that consists of a single large multifunctional polypeptide that catalyzes the steps in fatty acid elongation.Citation95

The FAS II enzyme enoyl-acetyl carrier protein reductase (ENR), which catalyzes the last step in fatty acid elongation, has been the focus of a number of drug development efforts. ENR is inhibited by the antibacterial compound triclosan, which inhibits the in vitro growth of T. gondii at low micromolar to nanomolar concentrations.Citation96,Citation97 Triclosan has a very low solubility in water and poor oral bioavailability and a number of triclosan derivatives have been synthesized to increase potency and solubility.Citation98Citation100 The most promising triclosan derivatives are described by Stec et al;Citation98 compound 16c of this series had an in vitro IC50 of 250 nM compared to 3 µM for triclosan. Compound 16c also demonstrated improved solubility over triclosan, with a computational log P of 3.9 versus 5.5 for triclosan. Compound 16c, when given at an IP dose of 75 mg/kg/day for 2 days starting on the day of infection, decreased T. gondii proliferation in vivo compared to vehicle controls. Doses of ≤50 mg/kg were not effective.

Samuel et alCitation101 demonstrated delivery of fluorescently labeled triclosan-octoarginine conjugates to intracellular and extracellular tachyzoites as well as encysted bradyzoites. Fluorescent triclosan-octoarginine conjugates were observed to localize around but not clearly enter the apicoplast. However, incubation of the RH strain in cell culture with a hydrolyzable triclosan-octoarginine conjugate resulted in concentration-dependent inhibition of T. gondii growth with maximum effect seen at 12 µM. Furthermore, administration of 40 mg/kg/day triclosan-octoarginine conjugate IP to mice infected IP with the RH strain reduced parasite counts over threefold after 5 days of treatment, compared to treatment with vehicle or unconjugated triclosan.

In another approach to improving the drug properties of triclosan, El-Zawawy et alCitation102 created triclosan-loaded liposomes and compared their efficacy against triclosan in a murine model of acute toxoplasmosis. Liposomal triclosan at an oral dose of 100 mg/kg/day reduced the parasite burden by 96% in peritoneal fluid compared to a reduction of 74% with standard triclosan at an oral dose of 150 mg/kg/day. There was no evidence of drug toxicity in uninfected controls treated with the same doses.

The benzimidazoles are a class of compounds known to target bacterial ENRs.Citation103 These compounds inhibit in vitro growth of T. gondii with IC50s in the low micromolar range.Citation104 Based on co-crystallization studies, these compounds do not seem to bind TgENR tightly, raising questions as to their primary mode of action.

Another validated drug target in the FAS II pathway is the enzyme β-ketoacyl-acyl carrier protein synthase, which is inhibited by thiolactomycin, a naturally occurring thiolactone.Citation105 Martins-Duarte et alCitation106 tested eight thiolactomycin analogs against RH strain T. gondii growing in LCC-MK2 cell culture and noted IC50s between 1.6 and 29.4 µM. Electron microscopy of treated parasites revealed swollen mitochondrial, enlarged Golgi complex cisternae, and incomplete separation of dividing daughter cells.

Fluoroquinolone derivatives

The fluoroquinolones are inhibitors of DNA gyrase and DNA topoisomerase IV widely used in human and veterinary medicine as antibacterial agents.Citation107Citation109 Trovafloxacin was found to have activity against T. gondii;Citation110 however, this drug is no longer used because of the risk of liver failure. Subsequent studies have examined fluoroquinolone derivatives and veterinary fluoroquinolones against T. gondii. Although the mechanism of action of fluoroquinolones against T. gondii is unknown, they are presumed to inhibit DNA synthesis in the apicoplast.

The veterinary fluoroquinolone enrofloxacin was found to have a modest protective effect against vertical transmission of the apicomplexan parasite Neospora caninum in a mouse model.Citation111 When tested against in vitro replication of T. gondii in human foreskin fibroblast (HFF) cell culture, enrofloxacin at 25 µg/mL reduced the number of cells infected by 59% versus an untreated control.Citation112 It was more effective than sulfadiazine at 100 µg/mL, which reduced the number of infected cells by 27%. Enrofloxacin also demonstrated efficacy in a murine model of infection with the ME49 strain, reducing brain cysts in Calomys callosus by 68%, similar to sulfadiazine (79%).

A number of fluoroquinolone derivatives have been synthesized in an effort to create more potent inhibitors of T. gondii.Citation113Citation115 Khan et alCitation113 evaluated six compounds with in vitro IC50s below that of trovafloxacin. Analogs 1 and 2 of this series both showed IC50s of 530 nM, compared to 2.93 µM for trovafloxacin. Dubar et alCitation116 synthesized several derivatives of ciprofloxacin and noted in vitro IC50s of 420 nM for compound 2, an ester prodrug, and 460 nM for compound 5, an adamantyl derivative. Neither compound was observed to be toxic to host cells at 30 µM. In a murine model of acute infection, compound 2 at 50 mg/kg/day orally for 7 days improved 60-day mortality to 18% versus 0% for ciprofloxacin-treated controls.Citation117 In the same model, compound 5 at 100 mg/kg/day orally for 7 days improved 60-day mortality to 25%.

Histone acetyltransferase/histone deacetylase inhibitors

The epigenetic control of gene expression by post-translational modification of histone proteins is a key process by which many organisms including T. gondii modulate transcription. Acetylation of conserved histone lysine residues by histone acetyltransferases (HATs) creates the post-translation modification, generally increasing transcription of the target gene. Conversely, de-acetylation by histone deacetylases (HDACs) removes the modification, generally decreasing the transcription of the target gene. The multiple stages in the T. gondii life cycle require significant changes in gene expression, and several groups have suggested developing novel therapeutic compounds that target epigenetic modification.Citation118,Citation119

The cyclic tetrapeptide FR235222 causes hyper-acetylation of histone H4 in T. gondii, inhibits growth of the parasite with an in vitro EC50 of 7.6 nM, and is associated with the conversion from the tachyzoite to bradyzoite stage.Citation120 Electron micrographs of bradyzoites formed during treatment with FR235222 show altered morphology and multiple nuclei. T. gondii resistant to FR235222 has T99A and T99I mutations in the HDAC TgHDAC3. Introducing either mutation into the parental strain reproduced the FR235222-resistant phenotype, demonstrating that TgHDAC3 is the target of FR235222. Interestingly, T99 is part of a two amino acid insertion (A98T99) that is unique to the members of the apicomplexan HDAC3 family, making this enzyme an attractive drug target.

Treatment of bradyzoites with FR235222 prevents differentiation into tachyzoites. Pretreated bradyzoites are incapable of infecting either an HFF monolayer in vitro or infecting mice.Citation121 However, host cell toxicity was a concern for FR235222 in that it inhibited HFF cells at an IC50 of 128 nM. Two derivatives of FR235222, W363 and W399, display IC50s against T. gondii equivalent to that of the parent compound but less toxic against HFF cells (HFF IC50:W363 =632 nM and W399 =539 nM).

A recently described HAT inhibitor is garcinol, a polyiso prenylated benzophenone derivative extracted from the kokum fruit (Garcinia indica) that targets the HAT TgGCN5b.Citation122 TgGCN5b is essential for tachyzoite replication.Citation123 T. gondii exposed to garcinol showed decreased transcription of genes regulated by TgGCN5b. Compared to other histone deacetyl transferase inhibitors the in vitro IC50 of garcinol was less potent at 1.8 µM.

N-benzoyl-2-hydroxybenzamides

In an effort to identify novel leads effective against T. gondii, Fomovska et alCitation124 screened a library of 6,811 synthetic compounds. The most promising compound to emerge from this screen, MP-IV-1, was found to have an IC90 of 31 nM against RH strain tachyzoites and no observed toxicity against host cells at 10 µM. A chemically diverse series of MP-IV-1 derivatives were synthesized and evaluated. Of these, the compound QQ-437 was the most potent with an in vitro IC90 of 16 nM and no apparent toxicity to host cells at 250 nM. Treatment with MP-IV-1 at a dose of 50 mg/kg/day IP reduced RH strain peritoneal fluid parasite counts. In an experiment using type II Prugniaud strain, QQ-437 at a dose of 20 mg/kg/day decreased parasite burden compared to MP-IV-1.

Using an insertional mutagenesis library, the authors identified four clones, all with insertions in the gene coding for adaptin-3β. Although little is known about adaptin-3β in T. gondii, the related protein adaptin-1 is known to be involved in sorting proteins from the Golgi complex to rhoptries.Citation125 Rhoptries, along with micronemes and dense granules, are secretory organelles found in T. gondii whose biogenesis requires correct trafficking of proteins from the Golgi complex.Citation126 Electron microscopy of parasites treated with either MP-IV-1 or QQ-437 demonstrated distortion of micronemes, dense granules, and rhoptries along with the absence of acidocalcisome, another secretory organelle. These results demonstrate that the N-benzoyl-2-hydroxybenzamides are potent inhibitors of secretory processes in T. gondii.

Macrolide derivatives

The apicoplast is a plastid organelle present in most of the members of the phylum Apicomplexa acquired from endosymbiosis.Citation127 The T. gondii apicoplast is the site of several essential metabolic pathways, many of which contain potential drug targets.Citation128 Molecules that inhibit protein synthesis in the apicoplast have been shown to be effective agents against T. gondii. Clindamycin inhibits the bacterial 50S ribosomal subunit and is used clinically as an antibacterial and for the treatment of toxoplasmosis. The macrolides erythromycin and azithromycin also target the 50S subunit and are routinely prescribed for the treatment of bacterial infections. Azithromycin prevents death from acute toxoplasmosis in a murine modelCitation129 and has several advantageous properties as a drug including high oral bioavailability and a long half-life. Similar to clindamycin, azithromycin causes a delayed death phenotypeCitation130 in which parasites are only modestly inhibited during the first round of replication, but all daughter parasites are severely impaired in their ability to replicate further, even after the removal of azithromycin.Citation131

Lee et alCitation132 synthesized a series of erythromycin and azithromycin analogs with the goal of improving antiparasitic efficacy. Compound 11 of their series is an azithromycin derivative in which the ring nitrogen was alkylated with a benzyl substituent. Compound 11 had an IC50 of 2 µM. Compound 11 displayed a similar delayed death phenotype as azithromycin with an IC50 of 5 µM against the first cycle of replication and an IC50 of 500 nM against the second cycle. Interestingly, in contrast to azithromycin, compound 11 significantly reduced the number of parasitophorous vacuoles formed on initial infection, suggesting an alternate mode of action.

Pantothenate synthetase inhibitors

All organisms that require coenzyme A and acyl carrier protein have pathways for synthesizing or acquiring these cofactors from pantothenate (vitamin B5). In plants, fungi, and bacteria, pantothenate is synthesized de novo from pyruvate via a conserved three-enzyme pathway;Citation133 however, animals must acquire pantothenate through their diet or from gastrointestinal bacteria. By searching the T. gondii genome for orthologs of the pantothenate synthesis pathway, Mageed et alCitation134 identified three conserved genes coding for de novo pantothenate biosynthesis in T. gondii. Among these genes is the terminal enzyme in the pathway, pantothenate synthetase that converts pantoate to pantothenate. Consistent with this finding, T. gondii did not exhibit a requirement for exogenous pantothenate when grown in culture.

Mageed et alCitation134 tested a series of pantothenate synthetase inhibitors originally developed for Mycobacterium tuberculosisCitation135 and found that two compounds, SW413 and SW404, inhibited in vitro growth of T. gondii with IC50s at 20 and 130 nM, respectively. Supplementing the growth media with 50 mg/L pantothenate decreased drug susceptibility to SW404 216-fold. In silico modeling placed SW404 in the pantoate-binding site of pantothenate synthetase and predicted binding to two conserved glutamine residues of this enzyme.

These results suggest that de novo pantothenic acid biosynthesis is an attractive target for drug development due to the lack of a homologous pathway in the human host. The rapid metabolism of free pantothenate by host cells to coenzyme A would likely prevent T. gondii from scavenging pantothenate from the host and overcoming pantothenate synthetase inhibition; however, this would need to be verified by in vivo experiments.

Peptide-conjugated phosphorodiamidate morpholino oligomers (PMOs)

PMOs are synthetic oligomers that bind complementary mRNA sequences and interfere with gene expression.Citation136 PMOs are potentially a highly specific means of disrupting the translation of key parasitic proteins. The challenge for developing effective PMOs is delivering the PMO to the target. Using a similar approach to that of Samuel et al,Citation105 Lai et alCitation137 conjugated various PMOs to arginine octomers, creating peptide-conjugated PMOs (PPMOs). The authors demonstrated specific knockdown of T. gondii DHFR, enoyl-acyl carrier protein reductase, and the transcription factor AP2XI-3, a key regulator of bradyzoite differentiation. Knocking down expression of each protein with 3–5 µM of the targeted PMO partially inhibited the replication of RH strain tachyzoites without toxicity to the host HFF cells at 20 µM. Growth of parasites treated with an anti-DHFR PPMO was rescued by supplementation with exogenous folic acid.Citation137 When a dose of 12.5 mg/kg was given IP for 2 days, the anti-DHFR PPMO reduced parasite count by 83% after 96 h in mice infected with RH strain T. gondii compared to treatment with vehicle or an off-target PPMO. PPMOs targeting the 5′ end of the T. gondii cytochrome b gene partially inhibited T. gondii replication at 10 µM.Citation42 In principal, PPMOs could be extended to target many essential genes.Citation137,Citation138

4-(1H)-quinolones

The cytochrome bc1 complex (bc1) is a drug target in several apicomplexan pathogens, including T. gondii, P. falciparum and B. microti. The bc1 reduces cytochrome c as part of the electron transport chain and generates an electrochemical gradient by transferring protons to the intermembrane space. The bc1 also creates ubiquinone for pyrimidine biosynthesis. The bc1 Qo site oxidizes ubiquinol and the bc1 Qi site reduces ubiquinone. Atovaquone is a Qo-site inhibitor that is used as an alternate treatment and prophylactic for toxoplasmosis, and in drug-combination regimens for malaria and mild to moderate babesiosis.Citation37,Citation139 Qi site inhibitors are not currently in clinical use; however, pyridones, which are Qi site inhibitors, were advanced to human studies for malaria, but were found to have cardiotoxicity in rats and activity against human bc1.Citation140 The endochin-like-quinolone (ELQ) series of 4(1H)-quinolone-3-diarylethers are derived from endochin, target the Qi site and have been designed to avoid human bc1 inhibition.Citation141,Citation142

Endochin was initially investigated as an antimalarial drug in an avian model of malaria by Salzer et al in 1948.Citation143 Gingrich and Darrow later found endochin to be active against avian and murine toxoplasmosis in 1951.Citation144 A library of 4(1H)-quinolone-3-diarylethers was made to improve metabolic stability, solubility and the in vivo efficacy of endochin. ELQ-316 and ELQ-271 were found to be highly effective against acute and latent toxoplasmosis.Citation39 The in vitro IC50 values of ELQ-271 and ELQ-316 were 0.1 and 0.007 nM, respectively. The ED50 values of ELQ-271 and ELQ-316 were 0.14 and 0.08 mg/kg when administered orally against acute toxoplasmosis with the RH strain in mice. ELQ-271 and ELQ-316 reduced the cyst burden in a mouse model of latent infection with the ME49 strain by 76%–88%.Citation39 ELQ-271 was found to inhibit the human bc1 at 800 nM, whereas ELQ-316 did not inhibit the human bc1 and was not toxic to human fibroblasts or human hepatocarcinoma cells (HepG2) at 10 µM, the highest concentration tested.Citation141 Despite being highly efficacious, the bioavailability of ELQs was limited by crystallinity and a lack of aqueous solubility, which was viewed as a liability for further clinical development. Improved bioavailability was achieved by the synthesis of ethyl carbonate prodrugs in which the ethyl carbonate promoiety disrupts crystal lattice formation.Citation145 Like atovaquone, ELQ-316 has broad anti-apicomplexan activity and is also highly effective against P. falciparum and B. microti and offers a novel approach to prevent the development of resistance when combined with atovaquone for dual inhibition of the parasite bc1.Citation141,Citation146

The 4-(1H)-quinolone scaffold was also recently investigated by McPhillie et al.Citation42 A screen of 5,6,7,8- tetrahydroquinolin-4-one derivatives identified MJM170 as a lead compound that inhibits RH tachyzoites in vitro with an IC50 of 30 nM and EGS encysted bradyzoites with an IC50 of 4 µM. A 25 mg/kg daily dose given for 5 days IP reduced the acute infection in mice with RH and Prugniaud parasites, and a 12.5 mg/kg dose given for 17 days reduced ME49 brain cysts. Inhibition of Saccharomyces cerevisiae bc1 Qi-site mutants and co-crystallography with bovine bc1 indicate that MJM170 binds to the bc1 Qi site.

The compound 1-hydroxy-2-dodecyl-4(1H)quinolone (HDQ), a structural analog of ubiquinone, has been identified as an inhibitor of type II NADH dehydrogenase isolated from the yeast Yarrow lipolitica, S. cerevisiae bc1 Qi site and T. gondii dihydroorotate dehydrogenase.Citation147Citation149 The electron transport chain of T. gondii contains a single-component type II NADH dehydrogenase instead of the multisubunit type I NADH dehydrogenase found in mammals.Citation150,Citation151 Hegewald et alCitation152 investigated HDQ in T. gondii by generating RH-strain mutants resistant to HDQ. Sequencing of genes coding for ubiquinone-binding proteins showed a N302S point mutation in dihydroorotate dehydrogenase. Enzymatic studies with purified wild-type and mutant enzyme confirmed that HDQ inhibited dihydroorotate dehydrogenase. HDQ was shown to inhibit the in vitro growth of RH strain T. gondii with an IC50 of 3.7 nM.Citation153 Later workCitation154 explored the activity of four HDQ derivatives, two of which were more potent than the parent molecule with in vitro IC50s of 0.4 and 0.8 nM (compounds A and B, respectively). The in vivo efficacy of these compounds was examined in murine models of acute and chronic infection. Compounds A and B showed similar efficacy as atovaquone in controlling parasite replication in the peritoneal cavity, but HDQ was less effective than atovaquone. HDQ, compound A, and compound B were effective against parasite replication in liver and lung, but not as effective as atovaquone. In a model of chronic infection with the ME49 strain, compound B showed a trend toward reduced brain cyst burden; however, this result did not reach statistical significance. Similar to the above-mentioned 4-(1H)-quinolones, HDQ derivatives are limited by low aqueous solubility via π-stacking of aromatic rings as Pidathala et alCitation155 describe in their investigation of HDQ derivatives.

Ruthenium complexes

Ruthenium-based complexes have been investigated for various purposes including the treatment of cancer and bacterial infections.Citation156 Barna et alCitation157 examined 18 ruthenium-containing compounds for activity against T. gondii, and they found that two compounds, 16 and 18 of their series, had IC50s of 18.7 and 41.1 nM, respectively. Treated parasites displayed empty or lipid-filled inclusions after 12 h of incubation with compound 16, and by 36 h, the parasite cytoplasm was disorganized. Both compounds 16 and 18 are hydrolytically stable ruthenium phosphite complexes with hydrocarbon exteriors. The authors hypothesize that the hydrocarbon exterior facilitates movement across lipid bilayers while shielding the core of the molecule from nucleophilic attacks and subsequent degradation.

Salicylanilides

Fomovska et alCitation158 synthesized 39 derivatives of the antihelmintic drug niclosamide and examined their in vitro efficacy against RH strain tachyzoites. Six derivative compounds had IC50s below 250 nM, and of these, four were tested further to assess whether they killed the parasites or the parasites resumed replication after removal of the compound (compounds 3i, 3j, 7a, and 14a). Parasites resumed replication after removal of compound 3i. Parasites did not recover after removal of 3j, 7a, or 14a. Two compounds, 14a and 14b, were tested for in vivo efficacy by challenging mice with oral infection of either ME49 or TgGoatUS4 oocysts and then treating orally with 100 mg/kg/day, 25 mg/kg/day, or vehicle control. All the infected mice died, but a mild protective effect was seen on treated animals as they died on average 1 day later. No gross toxicities were noted in animals treated with 100 mg/kg of either compound. The mechanism of action of salicylanilides is unknown despite efforts using random insertional mutagenesis to identify a target.

Spiroindolones

The spiroindolone NITD609 was originally discovered in a drug screen for new antimalarial agentsCitation159 and later found to be effective against T. gondii in vitro and in vivo. The target of NITD609 in P. falciparum is a P-type cation-transporting adenosine triphosphatase, PfATP4. In T. gondii, NITD609 has an in vitro IC50 of 1 µM against RH strain tachyzoites. In vivo, NITD609 at an oral dose of 100 mg/kg/day for 5 days decreased the parasite burden by 90% in mice infected IP with RH strain parasites. Although the T. gondii ATP4 has a high degree of sequence homology to PfATP4, further work is needed to definitively identify the target of NITD609 in T. gondii.

Thiosemicarbazones

Hydroxyurea, or hydroxycarbamide, is an antineoplastic drug primarily used in the treatment of polycythemia vera as well as other myeloproliferative disorders. In mammalian cells, it acts by inhibiting ribonucleotide reductase,Citation160 which catalyzes the rate-limiting step in DNA synthesis. Inhibition of ribonucleotide reductase leads to cell cycle arrest at the G1/S juncture. de Melo et alCitation161 demonstrated that incubation of infected Vero cells in 4 mM hydroxyurea for 5 h interfered with intracellular parasite replication. Electron microscopy showed that hydroxyurea caused severe morphologic alterations to intracellular parasites followed by the elimination of parasites from parasitophorous vacuoles.

Hydroxyurea is limited by its relatively low affinity for ribonucleotide reductase as well as its short half-life in humans.Citation160 However, the thiosemicarbazones have higher affinity for ribonucleoside reductaseCitation162 and are active against P. falciparum.Citation163

Tenório et alCitation164 synthesized a series of thiosemicarbazones as well as the structurally related 4-thiazolidinones and screened the resulting compounds for activity. Compounds were found to have IC50s ranging from 80 to 500 µM based on microscopy of infected host cells. By comparison, both hydroxyurea and sulfadiazine were found to have IC50s of 100 µM in this assay. The authors did not note any morphologic alterations to the Vero cells at the highest compound concentration of 30 mM. Of all the compounds tested, 4-thiazolidinones with phenyl substituents on the N-3 position showed the lowest IC50s. With this in mind, de Aquino et alCitation165 made an additional set of 4-thiazolidinones with the phenyl group fixed at the N-3 position and a set of thiosemicarbazones with the phenyl at the N-4 position (4-arylthiazolidinones). The two compounds with the lowest IC50, 2i and 2k, were 4-arylthiazolidinones with IC50s of 50 µM against intracellular tachyzoites and 1 mM against the host Vero cells.

Liesen et alCitation166 continued this work by synthesizing additional 4-thiazolidinones and 1,3,4-thiadiazoles from acylthiosemicarbazides with imidazole substituents at their N-1 position. Several members of this series showed equal efficacy to compounds from previous series but with less toxicity toward the host cells. Dzitko et alCitation167 expanded on this series and examined how replacement of the imidazole ring with other heteroaryl rings might alter the activity. The most potent compound in their series 1g had an IC50 of 33.2 µg/mL (107 µM) against RH tachyzoites in L929 fibroblast culture as measured by the incorporation of [3H]uracil. The authors conducted in silico studies in which these molecules were docked against possible target enzymes; however, no clear target was identified.

Triazines

The anticoccidial triazine toltrazuril is widely used in the poultry industry for its activity against Eimeria spp. This drug is not used in humans due to its potential to cause developmental malformations in a rat model.Citation168 The major metabolite of toltrazuril is ponazuril. Mitchell et alCitation169 demonstrated that ponazuril reduced the proliferation of T. gondii grown in African green monkey kidney cells at a concentration of 1 mg/mL (2.2 µM). The authors also found that oral ponazuril (10 mg/kg/day for 10 days) given 6 days after subcutaneous infection with RH parasites reduced mortality at 8 weeks (5/5 control mice dead versus 3/5 treated). With a higher dose (20 mg/kg/day for 10 days) given 6 days post-infection, all treated mice survived to 8 weeks.

Triazole derivatives

Itraconazole, a member of the triazole family of antifungal agents, is known to have in vitro activity against T. gondii tachyzoites.Citation170 However, in vivo studies of its protective effect against mortality and brain cyst formation in a mouse model of infection with the ME49 strain failed to demonstrate efficacy.Citation171 In the same study, the authors showed that the related compound fluconazole reduced 45-day mortality from 50% in untreated controls to 5% when given at 20 mg/kg/day to mice infected IP with ME49. However, fluconazole treatment did not reduce the formation of brain cysts when compared to untreated animals.

The idea of optimizing triazoles for better antiparasitic activity is a particularly attractive one, as these compounds are well tolerated and have high oral bioavailability. Dzitko et alCitation172 synthesized two such compounds, namely 3-(thiophen-2-yl)-1,2,4-triazole-5-thione and 4-ethyl-3-(4-methyl-1,2,3-thiadiazol-5-yl)-1,2,4–5-thione. The first of these, which the authors describe as “s-triazole” showed in vitro activity against the RH strain of T. gondii grown in L929 mouse fibroblast culture. IC50s measured by incorporation of 3H-uracil and quantitative real-time PCR were 42.46 µg/mL (235.6 µM) and 37.33 µg/mL (207.1 µM), respectively. These compared favorably to those obtained for sulfadiazine by the same methods, which were 2,225.25 µg/mL (8.9 mM) by 3H-uracil incorporation and 1,217.62 µg/mL (4.86 mM) by quantitative real-time PCR. Cytotoxic effects on host cells were measured using the MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay and calculated as the concentration needed to cause a 30% maximal effect (CC30). The CC30 for s-triazole was 194.46 µg/mL (1.08 mM) and that of sulfadiazine was 537.06 µg/mL (2.15 mM), demonstrating a better therapeutic index for s-triazole as calculated by IC50/CC30.

The target of s-triazole and other triazole compounds in T. gondii is unclear. Through a literature search, Dzitko et alCitation172 identified six likely enzymes and attempted in silico docking of s-triazole. Out of three potential targets, the authors suggested that Protein Data Bank number 3AU9 (1-deoxy-d-xylulose-5-phosphate reductoisomerase) was the most likely, followed by 4M84 (calmodulin-domain protein kinase 1) and 3MB8 (purine nucleoside phosphorylase).

Tryptanthrin derivatives

Tryptanthrin (indolo[2,1-b]quinazoline-6,12-dione) is a compound found in several natural sources, among them the plant Isatis indigotica. Krivogorsky et alCitation173 examined the activity of tryptanthrin and derivative molecules in vitro. Several compounds, in particular derivatives with halogen substituents at the 8 position, had IC50s under 10 nM. Further workCitation174 investigated the structure-activity relationship of the 6-keto group by replacing it with oximes, hydrazones, or alcohols. The oxime and alcohol derivatives of 8-bromotrypanthrin had excellent activity, with IC50s of 3 nM (for the oxime, compound 6b) and 2 nM (for the alcohol, compound 15b). Cytotoxic effects were not seen at concentrations as high as 100 µM. The target of these compounds was not determined.

Acknowledgments

This work was supported by Career Development Award # BX002440 from the United States Department of Veterans Affairs Biomedical Laboratory Research and Development.

Disclosure

The authors report no conflicts of interest in this work.

References

  • TenterAMHeckerothARWeissLMToxoplasma gondii: from animals to humansInt J Parasitol20003012–131217125811113252
  • JonesJLAksteinRBHlavsaMCLopezASWilsonMHollandGNFollow-up of the 1977 Georgia outbreak of toxoplasmosisAm J Trop Med Hyg20169461299130027044565
  • VaudauxJDMuccioliCJamesERIdentification of an atypical strain of toxoplasma gondii as the cause of a waterborne outbreak of toxoplasmosis in Santa Isabel do Ivai, BrazilJ Infect Dis201020281226123320836703
  • BowieWRKingASWerkerDHOutbreak of toxoplasmosis associated with municipal drinking water. The BC Toxoplasma Investigation TeamLancet199735090721731779250185
  • PappasGRoussosNFalagasMEToxoplasmosis snapshots: global status of Toxoplasma gondii seroprevalence and implications for pregnancy and congenital toxoplasmosisInt J Parasitol200939121385139419433092
  • DubeyJPLagoEGGennariSMSuCJonesJLToxoplasmosis in humans and animals in Brazil: high prevalence, high burden of disease, and epidemiologyParasitology2012139111375142422776427
  • JonesJLKruszon-MoranDRiveraHNPriceCWilkinsPPToxoplasma gondii seroprevalence in the United States 2009–2010 and comparison with the past two decadesAm J Trop Med Hyg20149061135113924710615
  • KhanAJordanCMuccioliCGenetic Divergence of Toxoplasma gondii Strains Associated with Ocular Toxoplasmosis, BrazilEmerging Infectious Diseases200612694294916707050
  • DemarMHommelDDjossouFAcute toxoplasmoses in immunocompetent patients hospitalized in an intensive care unit in French GuianaClin Microbiol Infect2012187E221E23121958195
  • JonesJLHollandGNAnnual burden of ocular toxoplasmosis in the USAm J Trop Med Hyg201082346446520207874
  • Robert-GangneuxFDardeMLEpidemiology of and diagnostic strategies for toxoplasmosisClin Microbiol Rev201225226429622491772
  • CoccaroEFLeeRGroerMWCanACoussons-ReadMPostolacheTTToxoplasma gondii infection: relationship with aggression in psychiatric subjectsJ Clin Psychiatry201677333434127046307
  • SugdenKMoffittTEPintoLPoultonRWilliamsBSCaspiAIs Toxoplasma gondii infection related to brain and behavior impairments in humans? Evidence from a population-representative birth cohortPLoS One2016112e014843526886853
  • SutterlandALFondGKuinABeyond the association. Toxoplasma gondii in schizophrenia, bipolar disorder, and addiction: systematic review and meta-analysisActa Psychiatr Scand2015132316117925877655
  • RabaudCMayTAmielCExtracerebral toxoplasmosis in patients infected with HIV. A French National SurveyMedicine (Baltimore)19947363063147984082
  • GrantIHGoldJWRosenblumMNiedzwieckiDArmstrongDToxoplasma gondii serology in HIV-infected patients: the development of central nervous system toxoplasmosis in AIDSAIDS1990465195212386617
  • MeleAPatersonPJPrenticeHGLeoniPKibblerCCToxoplasmosis in bone marrow transplantation: a report of two cases and systematic review of the literatureBone Marrow Transplant20022969169812180115
  • MartinoRMaertensJBretagneSToxoplasmosis after hematopoietic stem cell transplantationClin Infect Dis2000311188119511073751
  • MartinoRBretagneSEinseleHEarly detection of Toxoplasma infection by molecular monitoring of Toxoplasma gondii in peripheral blood samples after allogeneic stem cell transplantationClin Infect Dis2005401677815614694
  • MartinoRBretagneSRoviraMToxoplasmosis after hematopoietic stem transplantation. Report of a 5-year survey from the Infectious Diseases Working Party of the European Group for Blood and Marrow TransplantationBone Marrow Transplant2000251111111410828877
  • GajurelKDhakalRMontoyaJGToxoplasma prophylaxis in haematopoietic cell transplant recipients: a review of the literature and recommendationsCurr Opin Infect Dis201528428329226098500
  • JonesJLRobertsJMToxoplasmosis hospitalizations in the United States, 2008, and trends, 1993–2008Clin Infect Dis2012547e58e6122267718
  • HoffmannSBatzMBMorrisJGJrAnnual cost of illness and quality-adjusted life year losses in the United States due to 14 foodborne pathogensJ Food Prot20127571292130222980013
  • McLeodRBoyerKKarrisonTOutcome of treatment for congenital toxoplasmosis, 1981–2004: the National Collaborative Chicago-Based, Congenital Toxoplasmosis StudyClin Infect Dis200642101383139416619149
  • CarmeBDemarMAjzenbergDDardeMLSevere acquired toxoplasmosis caused by wild cycle of Toxoplasma gondii, French GuianaEmerg Infect Dis200915465665819331765
  • KhanAAjzenbergDMercierAGeographic separation of domestic and wild strains of Toxoplasma gondii in French Guiana correlates with a monomorphic version of chromosome1aPLoS Negl Trop Dis201489e318225233228
  • DemarMAjzenbergDMaubonDFatal outbreak of human toxoplasmosis along the Maroni River: epidemiological, clinical, and parasitological aspectsClin Infect Dis2007457e88e9517806043
  • PorterSBSandeMAToxoplasmosis of the central nervous system in the acquired immunodeficiency syndromeN Engl J Med199232723164316481359410
  • KatlamaCDe WitSO’DohertyEVan GlabekeMClumeckNPyrimethamine-clindamycin vs. pyrimethamine-sulfadiazine as acute and long-term therapy for toxoplasmic encephalitis in patients with AIDSClin Infect Dis19962222682758838183
  • DannemannBMcCutchanJAIsraelskiDTreatment of toxoplasmic encephalitis in patients with AIDS. A randomized trial comparing pyrimethamine plus clindamycin to pyrimethamine plus sulfadiazine. The California Collaborative Treatment GroupAnn Intern Med1992116133431727093
  • BeraudGPierre-FrancoisSFoltzerACotrimoxazole for treatment of cerebral toxoplasmosis: an observational cohort study during 1994–2006Am J Trop Med Hyg200980458358719346380
  • YanJHuangBLiuGMeta-analysis of prevention and treatment of toxoplasmic encephalitis in HIV-infected patientsActa Trop2013127323624423707647
  • TorreDCasariSSperanzaFRandomized trial of trimethoprim-sulfamethoxazole versus pyrimethamine-sulfadiazine for therapy of toxoplasmic encephalitis in patients with AIDS. Italian Collaborative Study GroupAntimicrob Agents Chemother1998426134613499624473
  • TorresRAWeinbergWStansellJAtovaquone for salvage treatment and suppression of toxoplasmic encephalitis in patients with AIDS. Atovaquone/Toxoplasmic Encephalitis Study GroupClin Infect Dis19972434224299114194
  • ChirgwinKHafnerRLeportCRandomized phase II trial of atovaquone with pyrimethamine or sulfadiazine for treatment of toxoplasmic encephalitis in patients with acquired immunodeficiency syndrome: ACTG 237/ANRS 039 Study. AIDS Clinical Trials Group 237/Agence Nationale de Recherche sur le SIDA, Essai 039Clin Infect Dis20023491243125011941551
  • JacobsonJMHafnerRRemingtonJDose-escalation, phase I/II study of azithromycin and pyrimethamine for the treatment of toxoplasmic encephalitis in AIDSAIDS200115558358911316995
  • Panel on Opportunistic Infections in HIV-Infected Adults and Adolescents Guidelines for the prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from the Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America Available at http://aidsinfo.nih.gov/contentfiles/lvguidelines/adult_oi.pdfAccessed July 17, 2016
  • HarrellMCarvounisPECurrent treatment of toxoplasma retinochoroiditis: an evidence-based reviewJ Ophthalmol2014201427350625197557
  • DoggettJSNilsenAForquerIEndochin-like quinolones are highly efficacious against acute and latent experimental toxoplasmosisProc Natl Acad Sci U S A201210939159361594123019377
  • BenmerzougaICheckleyLAFerdigMTArrizabalagaGWekRCSullivanWJJrGuanabenz repurposed as an antiparasitic with activity against acute and latent toxoplasmosisAntimicrob Agents Chemother201559116939694526303803
  • AraujoFGHuskinson-MarkJGutteridgeWERemingtonJSIn vitro and in vivo activities of the hydroxynaphthoquinone 566C80 against the cyst form of Toxoplasma gondiiAntimicrob Agents Chemother19923623263301605598
  • McPhillieMZhouYEl BissatiKNew paradigms for understanding and step changes in treating active and chronic, persistent apicomplexan infectionsNature Publishing Group2016629179
  • WattsEZhaoYDharaAEllerBPatwardhanASinaiAPNovel approaches reveal that Toxoplasma gondii bradyzoites within tissue cysts are dynamic and replicating entities in vivoMBio201565e01155e0111526350965
  • GajurelKGomezCADhakalRVogelHMontoyaJGFailure of primary atovaquone prophylaxis for prevention of toxoplasmosis in hematopoietic cell transplant recipientsTranspl Infect Dis201618344645227016655
  • MeneceurPBouldouyreMAAubertDIn vitro susceptibility of various genotypic strains of Toxoplasma gondii to pyrimethamine, sulfadiazine, and atovaquoneAntimicrob Agents Chemother20085241269127718212105
  • AspinallTVJoynsonDHGuyEHydeJESimsPFThe molecular basis of sulfonamide resistance in Toxoplasma gondii and implications for the clinical management of toxoplasmosisJ Infect Dis2002185111637164312023770
  • DoliwaCXiaDEscotte-BinetSIdentification of differentially expressed proteins in sulfadiazine resistant and sensitive strains of Toxoplasma gondii using difference-gel electrophoresis (DIGE)Int J Parasitol Drugs Drug Resist20133354424533291
  • DoliwaCEscotte-BinetSAubertDSulfadiazine resistance in Toxoplasma gondii: no involvement of overexpression or polymorphisms in genes of therapeutic targets and ABC transportersParasite2013201923707894
  • McFaddenDCTomavoSBerryEABoothroydJCCharacterization of cytochrome b from Toxoplasma gondii and Q(o) domain mutations as a mechanism of atovaquone-resistanceMol Biochem Parasitol2000108111210802314
  • AraujoFGHuskinsonJRemingtonJSRemarkable in vitro and in vivo activities of the hydroxynaphthoquinone 566C80 against tachyzoites and tissue cysts of Toxoplasma gondiiAntimicrob Agents Chemother19913522932992024964
  • ReynoldsMGOhJRoosDSIn vitro generation of novel pyrimethamine resistance mutations in the Toxoplasma gondii dihydrofolate reductaseAntimicrob Agents Chemother20014541271127711257045
  • KimKA bradyzoite is a bradyzoite is a bradyzoite?Trends Parasitol2015311261061226515047
  • VidadalaRSRivasKLOjoKKDevelopment of an orally available and central nervous system (CNS)-penetrant Toxoplasma gondii calcium-dependent protein kinase 1 (TgCDPK1) inhibitor with minimal human ether-a-go-go-related gene (hERG) activity for the treatment of toxoplasmosisJ Med Chem201659136531654627309760
  • LüderCGKReichardUGrossUChapter 7 – Toxoplasma Animal Models and TherapeuticsToxoplasma Gondii (Second Edition)BostonAcademic Press2014217255
  • Paredes-SantosTCTomitaTYan FenMDevelopment of dual fluorescent stage specific reporter strain of Toxoplasma gondii to follow tachyzoite and bradyzoite development in vitro and in vivoMicrobes Infect2016181394726432517
  • SidikSMHuetDGanesanSMA genome-wide CRISPR screen in toxoplasma identifies essential apicomplexan genesCell2016166614231435.e1227594426
  • GajriaBBahlABrestelliJToxoDB: an integrated Toxoplasma gondii database resourceNucleic Acids Res200836Database issueD553D55618003657
  • NagamuneKBeattyWLSibleyLDArtemisinin induces calcium-dependent protein secretion in the protozoan parasite Toxoplasma gondiiEukaryotic Cell200762147215617766463
  • NagamuneKMorenoSNJSibleyLDArtemisinin-resistant mutants of Toxoplasma gondii have altered calcium homeostasisAntimicrob Agents Chemother2007513816382317698618
  • HolfelsEMcAuleyJMackDMilhousWKMcLeodRIn vitro effects of artemisinin ether, cycloguanil hydrochloride (alone and in combination with sulfadiazine), quinine sulfate, mefloquine, primaquine phosphate, trifluoperazine hydrochloride, and verapamil on Toxoplasma gondiiAntimicrob Agents Chemother199438139213968092843
  • D’AngeloJGBordónCPosnerGHYolkenRJones-BrandoLArtemisinin derivatives inhibit Toxoplasma gondii in vitro at multiple steps in the lytic cycleJ Antimicrob Chemother20096314615018988681
  • SchultzTLHenckenCPWoodardLEA thiazole derivative of artemisinin moderately reduces Toxoplasma gondii cyst burden in infected miceJ Parasitol201410051652124524228
  • HenckenCPJones-BrandoLBordónCThiazole, oxadiazole, and carboxamide derivatives of artemisinin are highly selective and potent inhibitors of Toxoplasma gondiiJ Med Chem2010533594360120373807
  • DrakeMTClarkeBLKhoslaSBisphosphonates: mechanism of action and role in clinical practiceMayo Clin Proc20088391032104518775204
  • LingYLiZ-HMirandaKOldfieldEMorenoSNJThe farnesyl-diphosphate/geranylgeranyl-diphosphate synthase of Toxoplasma gondii is a bifunctional enzyme and a molecular target of bisphosphonatesJ Biol Chem2007282308043081617724033
  • MartinMBGrimleyJSLewisJCBisphosphonates inhibit the growth of Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondii, and Plasmodium falciparum: a potential route to chemotherapyJ Med Chem20014490991611300872
  • YardleyVKhanAAMartinMBIn vivo activities of farnesyl pyrophosphate synthase inhibitors against Leishmania donovani and Toxoplasma gondiiAntimicrob Agents Chemother20024692993111850291
  • JohnsonSMMurphyRCGeigerJADevelopment of Toxoplasma gondii calcium-dependent protein kinase 1 (TgCDPK1) inhibitors with potent anti-toxoplasma activityJ Med Chem20125552416242622320388
  • WinzerPMullerJAguado-MartinezAIn vitro and in vivo effects of the bumped kinase inhibitor 1294 in the related cyst-forming apicomplexans Toxoplasma gondii and Neospora caninumAntimicrob Agents Chemother201559106361637426248379
  • LouridoSShumanJZhangCShokatKMHuiRSibleyLDCalcium-dependent protein kinase 1 is an essential regulator of exocytosis in ToxoplasmaNature2010465729635936220485436
  • OjoKKLarsonETKeylounKRToxoplasma gondii calcium-dependent protein kinase 1 is a target for selective kinase inhibitorsNat Struct Mol Biol201017560260720436472
  • MurphyRCOjoKKLarsonETDiscovery of potent and selective inhibitors of calcium-dependent protein kinase 1 (CDPK1) from C. parvum and T. gondiiACS Med Chem Lett20101733133521116453
  • SugiTKobayashiKTakemaeHIdentification of mutations in TgMAPK1 of Toxoplasma gondii conferring resistance to 1NM-PP1Int J Parasitol Drugs Drug Resist201339310124533298
  • SugiTKawazuSHorimotoTKatoKA single mutation in the gatekeeper residue in TgMAPKL-1 restores the inhibitory effect of a bumped kinase inhibitor on the cell cycleInt J Parasitol Drugs Drug Resist2015511825941623
  • LarsonETOjoKKMurphyRCMultiple determinants for selective inhibition of apicomplexan calcium-dependent protein kinase CDPK1J Med Chem20125562803281022369268
  • DoggettJSOjoKKFanEMalyDJVan VoorhisWCBumped kinase inhibitor 1294 treats established Toxoplasma gondii infectionAntimicrob Agents Chemother20145863547354924687502
  • LouridoSZhangCLopezMSOptimizing small molecule inhibitors of calcium-dependent protein kinase 1 to prevent infection by Toxoplasma gondiiJ Med Chem20135673068307723470217
  • SugiTKatoKKobayashiK1NM-PP1 treatment of mice infected with Toxoplasma gondiiJ Vet Med Sci201173101377137921685719
  • HuangWOjoKKZhangZSAR studies of 5-aminopyrazole- 4-carboxamide analogues as potent and selective inhibitors of CDPK1ACS Med Chem Lett20156121184118926693272
  • ZhangZOjoKKJohnsonSMBenzoylbenzimidazole-based selective inhibitors targeting Cryptosporidium parvum and Toxoplasma gondii calcium-dependent protein kinase-1Bioorg Med Chem Lett201222165264526722795629
  • ZhangZOjoKKVidadalaRPotent and selective inhibitors of CDPK1 from T. gondii and C. parvum based on a 5-aminopyrazole-4-carboxamide scaffoldACS Med Chem Lett2013514044
  • MoineEDenevault-SabourinCDebierre-GrockiegoFA small-molecule cell-based screen led to the identification of biphenylimidazoazines with highly potent and broad-spectrum anti-apicomplexan activityEur J Med Chem20158938640025462254
  • MoineEDimier-PoissonIEnguehard-GueiffierCDevelopment of new highly potent imidazo[1,2-b]pyridazines targeting Toxoplasma gondii calcium-dependent protein kinase 1Eur J Med Chem20151058010526479029
  • PelphreyPMPopovVMJoskaTMHighly efficient ligands for dihydrofolate reductase from Cryptosporidium hominis and Toxoplasma gondii inspired by structural analysisJ Med Chem20075094095017269758
  • CanfieldCJMilhousWKAgerALPS-15: a potent, orally active antimalarial from a new class of folic acid antagonistsAm J Trop Med Hyg1993491211268352384
  • FidockDAWellemsTETransformation with human dihydrofolate reductase renders malaria parasites insensitive to WR99210 but does not affect the intrinsic activity of proguanilProc Natl Acad Sci U S A19979410931109369380737
  • MuiEJSchiehserGAMilhousWKNovel triazine JPC-2067-B inhibits Toxoplasma gondii in vitro and in vivoPLoS Negl Trop Dis200823e19018320016
  • SmithMHPloeghHLWeissmanJSRoad to ruin: targeting proteins for degradation in the endoplasmic reticulumScience20113341086109022116878
  • HarbutMBPatelBAYeungBKSTargeting the ERAD pathway via inhibition of signal peptide peptidase for antiparasitic therapeutic designProc Natl Acad Sci U S A2012109214862149123236186
  • JelenskaJCrawfordMJHarbOSSubcellular localization of acetyl-CoA carboxylase in the apicomplexan parasite Toxoplasma gondiiProc Natl Acad Sci U S A2001982723272811226307
  • RobertsFRobertsCWJohnsonJJEvidence for the shikimate pathway in apicomplexan parasitesNature19983938018059655396
  • WallerRFKeelingPJDonaldRGNuclear-encoded proteins target to the plastid in Toxoplasma gondii and Plasmodium falciparumProc Natl Acad Sci U S A19989512352123579770490
  • MagnusonKJackowskiSRockCOCronanJERegulation of fatty acid biosynthesis in Escherichia coliMicrobiol Rev1993575225428246839
  • McFaddenGIRoosDSApicomplexan plastids as drug targetsTrends Microbiol1999732833310431206
  • WakilSJStoopsJKJoshiVCFatty acid synthesis and its regulationAnnu Rev Biochem1983525375796137188
  • Martins-DuarteESCariasMVommaroRSuroliaNde SouzaWApicoplast fatty acid synthesis is essential for pellicle formation at the end of cytokinesis in Toxoplasma gondiiJ Cell Sci2016129173320333127457282
  • McLeodRMuenchSPRaffertyJBTriclosan inhibits the growth of Plasmodium falciparum and Toxoplasma gondii by inhibition of apicomplexan Fab IInt J Parasitol20013110911311239932
  • StecJFomovskaAAfanadorGAModification of triclosan scaffold in search of improved inhibitors for enoyl-acyl carrier protein (ACP) reductase in Toxoplasma gondiiChemMedChem201381138116023776166
  • ChengGMuenchSPZhouYDesign, synthesis, and biological activity of diaryl ether inhibitors of Toxoplasma gondii enoyl reductaseBioorg Med Chem Lett2013232035204323453069
  • MuenchSPStecJZhouYDevelopment of a triclosan scaffold which allows for adaptations on both the A- and B-ring for transport peptidesBioorg Med Chem Lett2013233551355523664871
  • SamuelBUHearnBMackDDelivery of antimicrobials into parasitesProc Natl Acad Sci U S A2003100142811428614623959
  • El-ZawawyLAEl-SaidDMossallamSFRamadanHSYounisSSTriclosan and triclosan-loaded liposomal nanoparticles in the treatment of acute experimental toxoplasmosisExp Parasitol2015149546425499511
  • HevenerKEMehboobSSuP-CDiscovery of a novel and potent class of F. tularensis enoyl-reductase (FabI) inhibitors by molecular shape and electrostatic matchingJ Med Chem20125526827922098466
  • WilkinsonCMcPhillieMJZhouYThe benzimidazole based drugs show good activity against T. gondii but poor activity against its proposed enoyl reductase enzyme targetBioorg Med Chem Lett20142491191624398298
  • JackowskiSMurphyCMCronanJERockCOAcetoacetyl-acyl carrier protein synthase. A target for the antibiotic thiolactomycinJ Biol Chem1989264762476292651445
  • Martins-DuarteÉSJonesSMGilbertIHAtellaGCde SouzaWVommaroRCThiolactomycin analogues as potential anti-Toxoplasma gondii agentsParasitol Int20095841141519698800
  • DrlicaKZhaoXDNA gyrase, topoisomerase IV, and the 4-quinolonesMicrobiol Mol Biol Rev1997613773929293187
  • PanXSFisherLMTargeting of DNA gyrase in Streptococcus pneumoniae by sparfloxacin: selective targeting of gyrase or topoisomerase IV by quinolonesAntimicrob Agents Chemother1997414714749021211
  • MartinezMMcDermottPWalkerRPharmacology of the fluoroquinolones: a perspective for the use in domestic animalsVet J2006172102816154368
  • KhanAASliferTAraujoFGRemingtonJSTrovafloxacin is active against Toxoplasma gondiiAntimicrob Agents Chemother199640185518598843293
  • GottsteinBRazmiGRAmmannPSagerHMüllerNToltrazuril treatment to control diaplacental Neospora caninum transmission in experimentally infected pregnant miceParasitology19991304148
  • BarbosaBFGomesAOFerroEAVNapolitanoDRMineoJRSilvaNMEnrofloxacin is able to control Toxoplasma gondii infection in both in vitro and in vivo experimental modelsVet Parasitol2012187445222281151
  • KhanAAAraujoFGBrightyKEGootzTDRemingtonJSAnti-Toxoplasma gondii activities and structure-activity relationships of novel fluoroquinolones related to trovafloxacinAntimicrob Agents Chemother1999431783178710390245
  • AnquetinGRouquayrolMMahmoudiNSynthesis of new fluoroquinolones and evaluation of their in vitro activity on Toxoplasma gondii and Plasmodium sppBioorg Med Chem Lett2004142773277615125930
  • AnquetinGGreinerJMahmoudiNDesign, synthesis and activity against Toxoplasma gondii, Plasmodium spp., and Mycobacterium tuberculosis of new 6-fluoroquinolonesEur J Med Chem2006411478149317000032
  • DubarFWintjensRMartins-DuarteÉSEster prodrugs of ciprofloxacin as DNA-gyrase inhibitors: synthesis, antiparasitic evaluation and docking studiesMed Chem Commun20112430436
  • Martins-DuarteÉSDubarFLawtonPCiprofloxacin Derivatives Affect Parasite Cell Division and Increase the Survival of Mice Infected with Toxoplasma gondiiPLoS ONE201510e0125705012572325950173
  • AndrewsKTHaqueAJonesMKHDAC inhibitors in parasitic diseasesImmunol Cell Biol201190667722124373
  • VanagasLJeffersVBogadoSSDalmassoMCSullivanWJJrAngelSOToxoplasma histone acetylation remodelers as novel drug targetsExpert Rev Anti Infect Ther20141011891201
  • BougdourAMaubonDBaldacciPDrug inhibition of HDAC3 and epigenetic control of differentiation in Apicomplexa parasitesJ Exp Med200920695396619349466
  • MaubonDBougdourAWongYSActivity of the histone deacetylase inhibitor FR235222 on Toxoplasma gondii: inhibition of stage conversion of the parasite cyst form and study of new derivative compoundsAntimicrob Agents Chemother2010544843485020713670
  • JeffersVGaoHCheckleyLALiuYFerdigMTSullivanWJJrGarcinol inhibits GCN5-mediated lysine acetyltransferase activity and prevents replication of the parasite Toxoplasma gondiiAntimicrob Agents Chemother2016602164217026810649
  • WangJDixonSETingL-MLysine acetyltransferase GCN5b interacts with AP2 factors and is required for Toxoplasma gondii proliferationPLoS Pathog201410e100383024391497
  • FomovskaAHuangQEl BissatiKNovel N-benzoyl-2-hydroxybenzamide disrupts unique parasite secretory pathwayAntimicrob Agents Chemother2012562666268222354304
  • NgôHMYangMPaprotkaKPypaertMHoppeHJoinerKAAP-1 in Toxoplasma gondii mediates biogenesis of the rhoptry secretory organelle from a post-Golgi compartmentJ Biol Chem20032785343535212446678
  • BreinichMSFergusonDJPFothBJA dynamin is required for the biogenesis of secretory organelles in Toxoplasma gondiiCurr Biol20091927728619217293
  • KöhlerSDelwicheCFDennyPWA plastid of probable green algal origin in Apicomplexan parasitesScience1997275148514899045615
  • SeeberFBiosynthetic pathways of plastid-derived organelles as potential drug targets against parasitic apicomplexaCurr Drug Targets Immune Endocr Metabol Disord2003329910912769782
  • AraujoFGGuptillDRRemingtonJSAzithromycin, a macrolide antibiotic with potent activity against Toxoplasma gondiiAntimicrob Agents Chemother1988327557572840017
  • PfefferkornERNothnagelRFBorotzSEParasiticidal effect of clindamycin on Toxoplasma gondii grown in cultured cells and selection of a drug-resistant mutantAntimicrob Agents Chemother199236109110961510399
  • FicheraMERoosDSA plastid organelle as a drug target in apicomplexan parasitesNature19973904074099389481
  • LeeYChoiJYFuHChemistry and biology of macrolide antiparasitic agentsJ Med Chem2011542792280421428405
  • WebbMESmithAGAbellCBiosynthesis of pantothenateNat Prod Rep20042169572115565250
  • MageedSNCunninghamFHungAWPantothenic acid biosynthesis in the parasite Toxoplasma gondii: a target for chemotherapyAntimicrob Agents Chemother2014586345635325049241
  • HungAWSilvestreHLWenSCiulliABlundellTLAbellCApplication of fragment growing and fragment linking to the discovery of inhibitors of Mycobacterium tuberculosis pantothenate synthetaseAngew Chem Int20094884528456
  • SummertonJWellerDMorpholino antisense oligomers: design, preparation, and propertiesAntisense Nucleic Acid Drug Dev1997731871959212909
  • LaiB-SWitolaWHEl BissatiKMolecular target validation, antimicrobial delivery, and potential treatment of Toxoplasma gondii infectionsProc Natl Acad Sci U S A2012109141821418722891343
  • ZhouYFomovskaAMuenchSLaiBSMuiEMcLeodRSpiroindolone That Inhibits PfATPase4 Is a Potent, Cidal Inhibitor of Toxoplasma gondii Tachyzoites In Vitro and In Vivo. AntimicrobAgents Chemother20145817891792
  • VannierEKrausePJHuman babesiosisN Engl J Med2012366252397240722716978
  • CapperMJO’NeillPMFisherNAntimalarial 4(1H)-pyridones bind to the Qi site of cytochrome bc1Proc Natl Acad Sci U S A2015112375576025564664
  • NilsenAMileyGPForquerIPDiscovery, synthesis, and optimization of antimalarial 4(1H)-quinolone-3-diarylethersJ Med Chem20145793818383424720377
  • AldayPHBruzualINilsenAGenetic Evidence for Cytochrome b Qi Site Inhibition by 4(1H)-quinolone-3-diarylethers and Antimycin in Toxoplasma gondiiAntimicrob Agents Chemother2016
  • SalzerWTAndersagHA new type of compound active against avian malariaChem Ber1948811219
  • GingrichWDarrowEThe effect of endochin on experimental toxoplasmosisAm J Trop Med Hyg195131121714799708
  • MileyGPPouSWinterRELQ-300 prodrugs for enhanced delivery and single-dose cure of malariaAntimicrob Agents Chemother20155995555556026124159
  • LawresLAGargAKumarVRadical cure of experimental babesiosis in immunodeficient mice using a combination of an endochin-like quinolone and atovaquoneJ Exp Med2016 Epub66
  • VallieresCFisherNAntoineTHDQ, a potent inhibitor of Plasmodium falciparum proliferation, binds to the quinone reduction site of the cytochrome bc1 complexAntimicrob Agents Chemother20125673739374722547613
  • EschemannAGalkinAOettmeierWBrandtUKerscherSHDQ (1-hydroxy-2-dodecyl-4(1H)quinolone), a high affinity inhibitor for mitochondrial alternative NADH dehydrogenase: evidence for a ping-pong mechanismJ Biol Chem20052803138314215533932
  • HegewaldJGrossUBohneWIdentification of dihydroorotate dehydrogenase as a relevant drug target for 1-hydroxyquinolones in Toxoplasma gondiiMol Biochem Parasitol2013190161523747278
  • VercesiAERodriguesCOUyemuraSAZhongLMorenoSNRespiration and oxidative phosphorylation in the apicomplexan parasite Toxoplasma gondiiJ Biol Chem199827331040310479813002
  • MeloAMPBandeirasTMTeixeiraMNew insights into type II NAD(P)H:quinone oxidoreductasesMicrobiol Mol Biol Rev20046860361615590775
  • HegewaldJGroßUBohneWIdentification of dihydroorotate dehydrogenase as a relevant drug target for 1-hydroxyquinolones in Toxoplasma gondiiMol Biochem Parasitol201319061523747278
  • SalehAFriesenJBaumeisterSGroßUBohneWGrowth inhibition of Toxoplasma gondii and Plasmodium falciparum by nanomolar concentrations of 1-hydroxy-2-dodecyl-4(1H)quinolone, a high-affinity inhibitor of alternative (type II) NADH dehydrogenasesAntimicrob Agents Chemother2007511217122217242151
  • BajohrLLMaLPlatteCIn vitro and in vivo activities of 1-hydroxy-2-alkyl-4(1H)quinolone derivatives against Toxoplasma gondiiAntimicrob Agents Chemother20105451752119884369
  • PidathalaCAmewuRPacorelBIdentification, design and biological evaluation of bisaryl quinolones targeting Plasmodium falciparum type II NADH: quinone oxidoreductase (PfNDH2)J Med Chem2012551831184322364416
  • BeckfordFAThessingJShaloskiMSynthesis and characterization of mixed-ligand diimine-piperonal thiosemicarbazone complexes of ruthenium(II): biophysical investigations and biological evaluation as anticancer and antibacterial agentsJ Mol Struct2011992394721552381
  • BarnaFDebacheKVockCAKusterTHemphillAIn vitro effects of novel ruthenium complexes in Neospora caninum and Toxoplasma gondii tachyzoitesAntimicrob Agents Chemother2013575747575423979747
  • FomovskaAWoodRDMuiESalicylanilide inhibitors of Toxoplasma gondiiJ Med Chem2012558375839122970937
  • RottmannMMcNamaraCYeungBKSSpiroindolones, a potent compound class for the treatment of malariaScience20103291175118020813948
  • GwiltPRTracewellWGPharmacokinetics and pharmacodynamics of hydroxyureaClin Pharmacokinet1998343473589592619
  • de MeloEJMayerhofferROde SouzaWHydroxyurea inhibits intracellular Toxoplasma gondii multiplicationFEMS Microbiol Lett2000185798210731610
  • LiuMCLinTSSartorelliACSynthesis and antitumor activity of amino derivatives of pyridine-2-carboxaldehyde thiosemicarbazoneJ Med Chem199235367236771433178
  • WalcourtALoyevskyMLovejoyDBGordeukVRRichardsonDRNovel aroylhydrazone and thiosemicarbazone iron chelators with anti-malarial activity against chloroquine-resistant and -sensitive parasitesInt J Biochem Cell Biol20043640140714687919
  • TenórioRPCarvalhoCSPessanhaCSSynthesis of thiosemicarbazone and 4-thiazolidinone derivatives and their in vitro anti-Toxoplasma gondii activityBioorg Med Chem Lett2005152575257815863319
  • de AquinoTMLiesenAPda SilvaREASynthesis, anti-Toxoplasma gondii and antimicrobial activities of benzaldehyde 4-phenyl-3-thiosemicarbazones and 2-[(phenylmethylene)hydrazono]-4-oxo-3-phenyl-5-thiazolidineacetic acidsBioorg Med Chem20081644645617905587
  • LiesenAPde AquinoTMCarvalhoCSSynthesis and evaluation of anti-Toxoplasma gondii and antimicrobial activities of thiosemicarbazides, 4-thiazolidinones and 1,3,4-thiadiazolesEur J Med Chem2010453685369120541294
  • DzitkoKPanethAPlechT1,4-Disubstituted thiosemicarbazide derivatives are potent inhibitors of Toxoplasma gondii proliferationMolecules2014199926994325010466
  • WoodwardKNToxicological Effects of Veterinary Medical Products in Humans2Cambridge, UKThe Royal Society of Chemistry2013
  • MitchellSMZajacAMDavisWLLindsayDSEfficacy of ponazuril in vitro and in preventing and treating Toxoplasma gondii infections in miceJ Parasitol20049063964215270113
  • Martins-DuarteÉDSde SouzaWVommaroRCItraconazole affects Toxoplasma gondii endodyogenyFEMS Microbiol Lett200828229029818371067
  • Martins-DuarteÉSLemgruberLde SouzaWVommaroRCToxoplasma gondii: fluconazole and itraconazole activity against toxoplasmosis in a murine modelExp Parasitol201012446646920045696
  • DzitkoKPanethAPlechTPaweczykJWgliskaLPanethPTriazole-based compound as a candidate to develop novel medicines to treat toxoplasmosisAntimicrob Agents Chemother2014587583758525288090
  • KrivogorskyBGrundtPYolkenRJones-BrandoLInhibition of Toxoplasma gondii by indirubin and tryptanthrin analogsAntimicrob Agents Chemother2008524466446918824607
  • KrivogorskyBNelsonACDouglasKAGrundtPTryptanthrin derivatives as Toxoplasma gondii inhibitors – structure-activity-relationship of the 6-positionBioorg Med Chem Lett2013231032103523321561