Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2016 Nov 21;60(12):7017–7034. doi: 10.1128/AAC.01176-16

Review of Experimental Compounds Demonstrating Anti-Toxoplasma Activity

Madalyn M McFarland a, Sydney J Zach a, Xiaofang Wang b, Lakshmi-Prasad Potluri c, Andrew J Neville a, Jonathan L Vennerstrom b, Paul H Davis a,
PMCID: PMC5118980  PMID: 27600037

Abstract

Toxoplasma gondii is a ubiquitous apicomplexan parasite capable of infecting humans and other animals. Current treatment options for T. gondii infection are limited and most have drawbacks, including high toxicity and low tolerability. Additionally, no FDA-approved treatments are available for pregnant women, a high-risk population due to transplacental infection. Therefore, the development of novel treatment options is needed. To aid this effort, this review highlights experimental compounds that, at a minimum, demonstrate inhibition of in vitro growth of T. gondii. When available, host cell toxicity and in vivo data are also discussed. The purpose of this review is to facilitate additional development of anti-Toxoplasma compounds and potentially to extend our knowledge of the parasite.

INTRODUCTION

Toxoplasma gondii, a common protozoan parasite, has the ability to infect nearly all warm-blooded animals, including humans, on all seven continents (13). This infection, while often asymptomatic in otherwise-healthy individuals, can cause severe disease or death in immunocompromised individuals, as well as severe congenital defects in prenatally infected infants. T. gondii is often acquired through the consumption of contaminated foods, either from undercooked meats or inadequately washed fruits and vegetables. Exposure to contaminated water is also a significant risk factor for infection, as contaminated water sources have been implicated in multiple outbreaks (46). Chemotherapy for treating toxoplasmosis is currently limited to the acute parasitic life stage (tachyzoite) of the infection and frequently consists of combination treatments, most often the antifolates pyrimethamine and sulfadiazine (7). Despite the ability to coadminister folinic acid orally with pyrimethamine, patients can suffer hematopoietic deficiencies, among other adverse effects, due to folate synthesis inhibition (8). Moreover, sulfadiazine has one of the highest rates of allergic reactions to an antibiotic reported in the United States, affecting 3 to 6% of the population (9, 10).

For women who become infected while pregnant, the treatment protocol is less well-defined; indeed, no regimen of treatment for expecting mothers is FDA approved (11). Often, congenitally infected infants are placed on an aggressive regimen of pyrimethamine and sulfadiazine for a period of 6 to 12 months immediately after birth, although the child remains at a high risk of later manifestations of neurological deficits regardless of the clinical symptoms presented at birth (12).

METHODS

In order to address these deficits, thousands of experimental compounds have been synthesized and investigated for activity against T. gondii. This work details the outcome of a comprehensive review of the literature, including articles accessed from PubMed with the following parameters: a publication date between 1 January 1980 and 28 June 2016; English as the primary language of the publication; containing the keywords “toxoplasm* AND (drug* or treatment*)”. A total of 5,504 items were filtered to identify primary literature sources that evaluated the in vitro or in vivo efficacy of compounds that were not derivatives of clinically available drugs used to treat toxoplasmosis (13); thus for the purpose of this review, they were considered “experimental compounds” due to their novelty and lack of clinical availability. Compounds with 50% inhibitory concentrations (IC50s) of >10 μM were not considered unless the compound had demonstrated efficacy in vivo. Additionally, compounds were excluded if the IC50 was determined based solely on the less-reliable enzyme-linked immunosorbent assay method (14). Clinically available drugs (13) and natural products (15) with activity against T. gondii are not considered in this review.

The experimental compounds in this review are generally divided by the methodology by which they were discovered, then by the predicted mode of action (MoA). The compounds identified as part of mid- or high-throughput screens are listed in Table 3, regardless of their MoA status. Those that have a known or suspected MoA are listed in Table 1, while those without a known or suspected MoA are listed in Table 2. All compounds described in the tables have been indexed numerically according to their order of appearance, with compound structures available in Tables S1 to S3 in the supplemental material.

TABLE 3.

Compounds with demonstrated activity against Toxoplasma gondii that were identified via various screen typesa

Screen type and compound no. Reference(s) IUPAC name Targeted protein Predicted or demonstrated MoA In vitro IC50 (μM) Host cell TD50 (μM) In vivo (mouse) therapeutic dose (mg/kg/day) In vivo toxicity (mg/kg/day)
Phenotypic screens
    67 104 Benzyl ((S)-1-oxo-1-(((S)-3-oxopent-4-en-2-yl)amino)-3-phenylpropan-2-yl)carbamate TgDJ-1 Prevents attachment and invasion ∼2–6 ND ND ND
    68 101 2-Methyl-3-(4-(4-(trifluoromethoxy)phenoxy)phenyl)quinolin-4(1H)-one Cytochrome bc1 complex Inhibits mitochondrial respiratory chain 0.0001 9.3 >1 >50
    69 101 6-Fluoro-7-methoxy-2-methyl-3-(4-(4-(trifluoromethoxy)-phenoxy)phenyl)- quinolin-4(1H)-one Cytochrome bc1 complex Inhibits mitochondrial respiratory chain 0.000007 >50 >0.33 >50
    70 195 Ethyl 6-fluoro-7-methoxy-3-(4-(4-(trifluoromethoxy)phenoxy)phenyl)quinoline-2-carboxylate Cytochrome bc1 complex Inhibits mitochondrial respiratory chain 5 >320 ND ND
    71 98 2-(4-Fluorophenyl)-3-(20-hydroxybiphen-3-yl)-6-(thien-3-yl)imidazo[1,2-b]pyridazine ND ND ∼0.61 >50 ND ND
    72 98 3-(20-Hydroxybiphen-3-yl)-2-tert-butyl-6-(thien-3-yl)imidazo[1,2-a]pyridine ND ND ∼0.08 >50 ND ND
    73 98 2-(2-Methoxyphenyl)-3-(2'-hydroxybiphen-3-yl)-6-(thien-3-yl)imidazo[1,2-b]pyridazine ND ND ∼0.36 ∼32.0 ND ND
    74 98 3-(20-Hydroxybiphen-3-yl)-2-(2-methoxyphenyl)-6-(thien-3-yl)imidazo[1,2-a]pyridine ND ND ∼0.27 >50 ND ND
    75 98 3-((Biphen-3-yl)-2-(2-methoxyphenyl)imidazo[1,2-a]pyridin-6-yl)methanol ND ND ∼0.63 ∼42.2 ND ND
    76 196 N-[(E)-(1-propan-2-ylbenzimidazol-2-yl)methylideneamino]-1H-benzimidazol-2-amine ND Inhibits motility and invasion ∼1.36 >10 ND ND
    77 196 (5E)-5-(2,4-dinitrophenoxy)iminoquinolin-8-one ND ND ∼1.34 >10 4.4 ND
    78 196 3-Benzyl-6-[1-(2-ethoxyphenyl)-5-methyltriazol-4-yl]-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazole ND ND ∼0.57 >10 ND ND
    79 196 4-N-Benzyl-2-N-(2-methylphenyl)quinazoline-2,4-diamine hydrochloride ND Inhibits motility ∼1.12 >10 ND ND
    80 99 N-4-Ethyl-benzoyl-2-hydroxybenzamide Adaptin-3β Inhibits secretion of micronemes, rhoptries, and dense granules I0.031b >10 50 ND
    81 100 N-[4-(Diethylamino)benzoyl]-2-hydroxybenzamide Adaptin-3β Inhibits secretion of micronemes, rhoptries, and dense granules 0.016b >10 20 ND
Targeted screens
    82 119 1-(tert-Butyl)-3-(3-methylbenzyl)-4-amino-1H-pyrazolo[3,4-d]pyrimidine TgCDPK1 Inhibits invasion, gliding motility, microneme secretion, and egress 0.11 ND 5 ND
    83 119 1-(tert-Butyl)-3-(3-chlorobenzyl)-4-amino-1H-pyrazolo[3,4-d]pyrimidine TgCDPK1 Inhibits invasion, gliding motility, microneme secretion, and egress 0.03 ND 5 ND
    84 119 3-(2,3-Dichlorobenzyl)-1-isopropyl-4-amino-1H-pyrazolo[3,4-d]pyrimidine TgCDPK1 Inhibits invasion, gliding motility, microneme secretion, and egress 0.25 ND 5 ND
    85 119 1-(tert-Butyl)-3-(3,5-difluorobenzyl)-4-amino-1H-pyrazolo[3,4-d]pyrimidine TgCDPK1 Inhibits invasion, gliding motility, microneme secretion, and egress 0.61 ND 5 ND
    86 120, 197 1-((1-Methylpiperidin-4-yl)methyl)-3-(6-ethoxynaphthalen-2-yl)- 4-amino-1H-pyrazolo[3,4-d]pyrimidine TgCDPK1 Inhibits invasion, gliding motility, microneme secretion, and egress 0.14 ND 40 >100
    87 113 2,4-Diamino-5-methyl-6-(2¢,6¢-dimethylphenylthio)pyrrolo[2,3-d]pyrimidine TgDHFR Growth inhibition 1.92 >260 50 ND
    88 113 2,4-Diamino-5-methyl-6-(2¢,6¢-dimethylphenylthio)pyrrolo[2,3-d]pyrimidine hydrochloride salt TgDHFR Growth inhibition 2.15 >260 50 ND
    89 115 2-Phenylthio-indole TgNTPase-I, TgNTPase-II Growth inhibition ∼7 >50 ND ND
    90 115 2-(2-Naphthalenylthio)-1H-indole TgNTPase-I, TgNTPase-II Growth inhibition ∼3.6 >50 ND ND
    91 115 2-(1-Naphthalenylthio)-1H-indole TgNTPase-I, TgNTPase-II Growth inhibition ∼3.2 >50 ND ND
Cheminformatic screens
    92 87 5-Nitroso-8-hydroxyquinoline ND Oxidative stress 0.08 ND ND ND
    93 124 3-[(E)-2-(1,3-Benzodioxol-5-yl)ethenyl]-1H-quinoxalin-2-one ROP18 Destruction of parasitophorous vacuole 0.002 ND ND ND
Drug repurposing screens
    94 126 N-(2-Ethoxyphenyl)-2-[4-(furan-2-carbonyl)piperazin-1-yl]acetamide ND ND 0.19 >30 ND ND
    95 126 N-[4-(Dibutylsulfamoyl)phenyl]furan-2-carboxamide ND ND 1.07 >30 ND ND
    96 126 7-Chloro-4-pyrrolidin-1-ylquinoline ND ND 1.49 >30 ND ND
    97 126 N',N'-Dimethyl-N-(2-phenylquinolin-4-yl)ethane-1,2-diamine ND ND 1.95 >30 ND ND
    98 126 6-Bromo-2-methyl-3-propyl-1H-quinolin-4-one ND ND 3.05 >30 ND ND
    99 126 N-[4-[(4-Ethylpiperazin-1-yl)methyl]phenyl]-1H-pyrrolo[3,2-h]quinoline-2-carboxamide ND ND 3.85 >30 ND ND
    100 126 2-(2-Methoxyanilino)-3-piperidin-1-ylnaphthalene-1,4-dione ND ND 4.54 >30 ND ND
    101 127 2-[(E)-2-(4-Ethoxyphenyl)ethenyl]-1,3-benzoxazin-4-one ND ND 0.27 >39.8 ND ND
    102 127 2-Amino-4-(3,5-ditert-butyl-4-hydroxyphenyl)-5-oxo-4,6,7,8-tetrahydrochromene-3-carbonitrile ND ND 1.33 >36.2 ND ND
    103 127 Methyl N-[(E)-[phenyl(pyridin-2-yl)methylidene]amino]carbamodithioate ND ND 1.63 >42.9 ND ND
    104 198 3-(4-Chlorophenyl)1-(1,1-dimethylethyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine ND ND 0.2 >5 ND ND
a

The lowest observed IC50s are reported in cases in which multiple values were found in the literature. Structure data for each compound in the table can be found in Table S3 in the supplemental material. In vivo therapeutic dose data refer to the best concentration tested in the literature for toxicity and efficacy in a model organism. ND, not determined.

b

The IC90 value (rather than IC50) is reported for this compound.

TABLE 1.

Compounds with known or suspected modes of action against Toxoplasma gondiia

MoA and compound class and no. Reference(s) In vitro IC50 Host cell TD50 In vivo survivability In vivo chronic infection Parasite burden In vivo toxicity
Interference with invasion or egress
    Pyrazolopyrimidines 24, 129, 130
        Compound 1 0.003 μM (24)
        Compound 2 Not established (129) 100,000 RH strain tachyzoites injected i.p.; treatment did not increase survival (129) 1,000 RH strain tachyzoites injected i.p.; treatment decreased parasite load in liver and lungs by ∼10- to 100-fold and in brain by <5-fold at 4 dpi (129)
        Compound 3 0.060 μM (130) >10 μM HepG2 and CRL-8155 cells (130) Unreported no. of ME49 cysts injected i.p.; treatment decreased cyst count by 88.7% (130) “High inoculum” of RH strain tachyzoites injected i.p.; 99% reduction in spleen tachyzoites; 95% reduction in brain tachyzoites (130) >500 mg/kg/day in mice (PK information available for rats, dogs, calves, monkeys) (130)
    Pyridinylpyrroles 19, 21, 22
        Compound 4 0.32–200 μM (19, 21, 22) >10 μM HFF cells (19, 21, 22) 1,000–3,000 RH strain tachyzoites injected i.p.; treatment increased survival by 90% (19) ND 1,000–3,000 RH strain tachyzoites injected i.p.; no parasites detected in brain, lung, spleen, or peritoneal fluid during treatment; at 5–10 days posttreatment, parasites detected in brain, lung, spleen (19) >50 mg/kg/day (19, 22)
    Benzophenones 20, 131, 132
        Compound 5 0.14 μM (131) 0.21 μM HFF cells, 5.89 μM HeLa cells (131) ND ND ND ND
    Benzoylbenzimidazoles 25
        Compound 6 >1 μM for all tested compounds >30 μM for all tested compounds ND ND ND ND
    Diaryl ureas 28
        Compound 7 <0.5 μM >10 μM HFF cells ND ND ND ND
    Dinitroanilines 27, 133
        Compound 8 0.036 μM (27) >0.5 μM HFF cells (27) ND ND ND ND
    5-Aminopyrazole-4-carboxamides 134
        Compound 9 0.089 μM >40 μM CLR-8155 cells ND ND 105 RH strain tachyzoites injected i.p.; no parasites detected in peritoneal fluid, >5-fold reduction in brain tissue >10 mg/kg/day
Inhibition of DNA synthesis
    6-Benzylthioinosines 42, 135140
        Compound 10 9.3 μM (136) >50 μM HFF cells (136) 200 RH or TgAK-3 strain tachyzoites injected i.p. increased survival by 2 days ND ND ND
        Compound 11 4.3 μM (136) >50 μM HFF cells (136) Increased survival by 2–3 days (136) ND ND >300 mg/kg (136)
        Compound 12 7.3 μM (136) >50 μM HFF cells (136) Increased survival by 2–3 days (136) ND ND >300 mg/kg (136)
    Triazines and 2,4-diaminopyrido[2,3-d]pyrimidines 37, 114, 141148
        Compound 13 0.058 μM (143) ND ND ND ND ND
        Compound 14 0.02 μM (37) ND ND ND 10,000 RH strain tachyzoites injected i.p.; 99% reduction in parasites in peritoneal fluid (37) ND
        Compound 15 ~0.05 μM (114) >100 μM HFF cells (114) ND ND 10,000 RH strain tachyzoites injected i.p.; significant reduction of tachyzoites in peritoneal fluid (114) ND
    Sulfonamides (exptl) 39, 149
        Compound 16 0.05 μM (39) ND ND ND ND ND
        Compound 17 ND (149) ND 5,000 RH strain tachyzoites injected i.p.; treatment group had 10–50% survival (149) ND 5,000 RH strain tachyzoites injected i.p. (mice that survived acute trial used for parasite burden assay); no parasites detected in heart or kidneys (149) ND
    2,4-Diaminopteridines 150
        Compound 18 0.077 μM ND ND ND ND ND
    Phthalazinone derivatives 41
        Compound 19 1 μM ND ND ND ND ND
    Urea derivatives 151
        Compound 20 0.402 μM ND ND ND ND ND
    1-[4-(4-Nitrophenoxy)phenyl]propane-1-one 152
        Compound 21 36.2 μM 67.0 μM HeLa cells ND ND 100,000 RH strain tachyzoites injected i.p.; 40% reduction in tachyzoite burden in peritoneal cavity at 4 dpi ND
Inhibition of steroid synthesis
    Bisphosphonates 53, 153158
        Compound 22 0.28 μM (154) >826 μM KB cells (154) 5 C56 strain bradyzoites administered orally; 80% survival (154) ND ND ND
        Compound 23 0.55 μM (154) >892 μM KB cells (154) 5 C56 strain bradyzoites administered orally; 80% survival (154) ND ND ND
    ACAT inhibitors 51
        Compound 24 ∼3 μM >100 μM HFF cells ND ND ND ND
    Aryloxyphenoxy derivatives 48, 159
        Compound 25 2.03 μM (55) >50 μM Vero cells (48) ND ND ND ND
    Azasterols 54, 55, 160
        Compound 26 0.12 μM (55) ND ND ND ND ND
    Quinuclidines 52
        Compound 27 0.19 μM >15 μM LLC-MK2 cells ND ND ND ND
Fatty acid synthesis inhibition
    Acylsulfonamides 63
        Compound 28 0.02 ± 0.07 μM >1,000 μM HFF cells ND ND ND ND
    Benzimidazoles 62
        Compound 29 2.5 μM <10 μM HFF or PC3-Luc cells ND ND ND ND
    Thiolactomycin analogues 60
        Compound 30 1.6 μM >15 μM LLC-MK2 cells ND ND ND ND
Inhibition of virulence factors or host interactions
    Pyridinylimidazoles 69, 70, 161
        Compound 31 0.8–5 μM (70) ND 1,000 RH strain tachyzoites injected i.p.; 40% survival in treatment group (70) ND ND ∼>60 mg/kg/day (70)
        Compound 32 ∼3 μM (69) ND ND 1,000 RH-GFP strain tachyzoites injected i.p.; significant reduction in peritoneal exudate (69) >10 mg/kg/day (69)
Inhibition of transcription
    Cyclic tetrapeptides 74, 75, 162
        Compound 33 0.01 μM (MIC) (75) ND ND ND ND ND
        Compound 34 0.0076 μM (74) 0.107 μM HFF cells (74) ND ND ND ND
    Hydroxamic acids 77
        Compound 35 0.039 μM >10 μM HS68 cells ND ND ND ND
    Garcinol 78
        Compound 36 1.79 μM >10 μM HFF cells ND ND ND ND
Inhibition of reproduction or differentiation
    Gossypol and derivatives 84
        Compound 37 5–10 μM ∼40 μM HFF cells ND ND ND ND
    Lactacystin 81
        Compound 38 2 μM (significant inhibition of intracellular tachyzoite replication) 2 μM (HFF cells showed little to no signs of toxicity) ND ND ND ND
    3-Bromopyruvate 163
        Compound 39 <10 μM >10 μM LLC-MK2 cells ND ND ND ND
Effects on ROS regulation
    Quinones 88, 164169
        Compound 40 0.10 μM (164) >10 μM HFF cells (164) 2,500 RH strain tachyzoites or 10 C56 strain bradyzoites given orally; 0–20% survival in treatment group (164) ND ND <100 mg/kg/day (164)
        Compound 41 0.11 μM (164) >10 μM HFF cells (164) 2,500 RH strain tachyzoites or 10 C56 strain bradyzoites given orally; 40–50% survival in treatment group (164) ND ND ∼100 mg/kg/day (164)
        Compound 42 ND (168) ND (Coadministered with sulfadiazine) 1,000 RH strain tachyzoites injected i.p., 70% survival in treatment group; when 10 EGS strain cysts given orally, 90% survival in treatment group (168) 5 P strain cysts given orally, cyst burden reduced ∼58% with treatment (168) ND <100 mg/kg/day (165)
    Quinolines 87, 170
        Compound 43 0.0786 μM (87) 3.4 μM HS68 cells (87) ND ND ND ND
    Alkaloids 86, 97, 169, 171177
        Compound 44 0.0007 μM (173) 0.001 μM THP-1 cells (173) ND ND ND ND
        Compound 45 0.0007 μM (173) 0.392 μM THP-1 cells (173) ND ND ND ND
    Cationic dyes 85
        Compound 46 0.26 μM 0.55 μM mouse peritoneal macrophages ND ND ND ND
a

Individual compound information is listed with the index number assigned to the specific molecule's structure. For cases in which multiple IC50 values were reported in the literature, the lowest observed IC50 is reported. Anti-Toxoplasma compounds are categorized by MoA and grouped by structure. Structures for each compound can be found in Table S1 in the supplemental material. The term in vivo survivability assay data refer to experiments where model organisms (mice) were exposed to a lethal infectious dose of parasite, often via i.p. injection. The strain and dose of the parasite and the recipient host differed between studies, making comparisons between studies problematic. In vivo chronic infection and parasite burden data refer to tissue or fluid counts of parasites isolated from a host following a nonlethal infection or bradyzoite cyst burden in brain tissue (most often determined via PCR). ND, not determined; dpi, days postinfection; HFF, human foreskin fibroblasts.

TABLE 2.

Compounds without known modes of action against Toxoplasma gondiia

Compound class and no. Reference(s) In vitro IC50 Host cell TD50 In vivo survivability In vivo chronic infection Parasite burden In vivo toxicity
3-[{2-((E)-furan-2-ylmethylene)hydrazinyl}methylene]-1,3-dihydroindol-2-one (ATT-5126) 152
    Compound 47 19.7 μM 35.4 µM HeLa cells ND ND 10,000 RH strain tachyzoites injected i.p.; 19% reduction in tachyzoite burden in peritoneal cavity ND
6-Trifluoromethyl-2-thiouracil (KH-0562) 152
    Compound 48 32.2 μM 56.3 μM HeLa cells ND ND 10,000 RH strain tachyzoites injected i.p.; 24% reduction in tachyzoite burden in peritoneal cavity ND
Diamidines 178, 179
    Compound 49 0.03 μM (179) >2 μM HFF cells (179) ND ND ND ND
Metal and metal complexes 93, 180, 181
    Compound 50 0.0187 μM (180) 2.4 μM HFF cells (180) ND ND ND ND
    Compound 51 ND (93) ND ND ND 3,500 RH strain tachyzoites injected i.p.; group treated 4 days preinfection had ∼45%–50% reduced organ burdens; group treated 4 days postinfection had ∼86% reduced spleen burden (93) >200 μg/ml (93)
    Compound 52 3.6 μM (181) >200 μM LLC-MK2 cells (181) ND ND ND ND
Resorcinarenes 182
    Compound 53 ND 4,239 μM RAW 264.7 cells ME49 strain, unspecified no. or life stage of parasites; led to 50% increase in survival in treatment group 25 ME49 strain bradyzoites delivered orally; no reduction in brain cysts in treatment group ND >500 mg/kg
Semisynthetic artemisinins 90, 183187
    Compound 54 108 μM (90) ND 1,000,000 PRU-Luc-GFP strain tachyzoites injected i.p.; 60% survival in treatment group (90) (Coadministered with sulfadiazine until 23dpi) 1,000,000 PRU-Luc-GFP strain tachyzoites injected i.p.; significant reduction in brain cysts in treatment group (90) ND ND
    Compound 55 ND (186) ND 50 RH strain tachyzoites injected i.p.; treatment increased survival by 6–7 days (186) 18 ME49 strain bradyzoites injected i.p.; ∼40% reduction of cyst burden in treatment group (186) ND <10 mg/kg (186)
    Compound 56 0.25 μM >320 μM HFF cells ND ND ND ND
Aculeatins 188
    Compound 57 0.173 μM 0.173 μM K562 cells ND ND ND ND
Clodinafop and derivatives 189
    Compound 58 10 μM led to 70% growth inhibition; IC50 not established >400 μM HFF cells ND ND ND ND
Indirubin analogues 174
    Compound 59 0.18 μM 20 μM HFF cells ND ND ND ND
Quinones (exptl) 190, 191
    Compound 60 1.74 μM (191) >6 μg/ml THP-1 cells (191) ND ND ND ND
    Compound 61 3.37 μM (191) >6 μg/ml THP-1 cells (191) ND ND ND ND
    Compound 62 8.36 μM led to 96.5% growth inhibition; IC50 not established (190) >2 μg/ml in THP-1 cells (190) ND ND ND ND
    Compound 63 8.73 μM led to 92% growth inhibition; IC50 not established (190) >2 μg/ml in THP-1 cells (190) ND ND ND ND
Thiazolidinones 192
    Compound 64 0.9 μM 35 μM HFF cells ND ND ND ND
Thioureides 193
    Compound 65 0.3 μM No noted effect on HFF or Caco2 cells; exact numbers not reported ND ND ND ND
Indole-1,2-diones 194
    Compound 66 0.3 μM 6.4 μM HFF cells ND ND ND ND
a

Individual compound information is listed with the index number assigned to the specific molecule's structure. For cases in which multiple IC50 values were found in the literature, the lowest observed IC50 is reported. Anti-Toxoplasma compounds are grouped by structure. Structures for each compound can be found in Table S2 in the supplemental material. The in vivo survivability assay data refer to experiments where model organisms (mice) were exposed to a lethal infectious dose of parasite, often via i.p. injection. The strain and dose of the parasite and the recipient host differed between studies, making comparisons between studies problematic. In vivo chronic infection and parasite burden data refer to the tissue or fluid counts of parasites isolated from a host following a nonlethal infection or bradyzoite cyst burden in brain tissue (most often determined via PCR). ND, not determined; dpi, days postinfection; HFF, human foreskin fibroblasts.

For promising anti-Toxoplasma compounds, assessment of their ability to control T. gondii growth is a key step in drug development. Traditionally, successful compounds demonstrate the following: an IC50 under 10 μM, except in cases where the compound is used in combination with another compound; a high therapeutic index; and an ability to increase survival of an infected host animal after infection with a lethal dose of T. gondii (13). Promising compounds also ideally demonstrate marked decreases in brain and muscle parasite cyst (bradyzoite) counts, a measure of chronic infection; however, this has not been established to be clinically significant. As more is learned about the potentially deleterious effects of chronic infections, this may become a greater consideration (16). While few reviewed compounds have absorption, distribution, metabolism, and excretion (ADME) data, a lead compound should demonstrate favorable pharmacokinetic characteristics to avoid a late-stage failure during development (17).

COMPOUNDS WITH PROPOSED MODES OF ACTION

Interference with invasion or egress.

Host cell invasion and egress of T. gondii are two distinct, complex, vital processes for the survival of the parasite. During acute infection, T. gondii tachyzoites require invasion of a host cell, formation of a parasitophorous vacuole, replication, and egress. Invasion is thought to be conducted through the formation of a junction between the apical end of the parasite and the host cell plasma membrane (18). The parasite uses an actin-myosin motor system to enter the host cell while surrounding itself in the newly formed vacuole or membrane arising from the host plasma membrane. Examples of compounds that specifically target invasion are pyridinylpyrroles (19) and benzophenones (20), which target cyclic GMP-dependent kinase, a parasite protein implicated in the gliding motility and microneme adhesion of T. gondii and other coccidians to the host cell plasma membrane (21) (Table 1). The most potent of these, pyridinylpyrrole (compound 4), had an in vitro IC50 of 0.32 μM, and increased the survival rate of mice infected with a lethal dose of parasites by 90% when the drug was administered twice a day at 50 mg/kg of body weight (19, 21, 22).

T. gondii remains in the parasitophorous vacuole for the duration of its asexual reproduction and must egress in order to maintain virulence. Egress, a less-well-understood process, requires the parasite to pass through both the established parasitophorous vacuole and the host plasma membrane (18). While this cycle has been demonstrated to be potassium and calcium dependent and may involve host coordination, specific mechanisms are not known (23). An important parasite protein for both invasion and egress, calcium-dependent protein kinase 1 (TgCDPK1), has been shown to be targeted by pyrazolopyrimidines (24) and benzoylbenzimidazoles (25) (Table 1). This parasite target is part of an important signaling pathway involved in gliding, a required motion for T. gondii to cross the host cell's plasma membrane (26). Other compounds that affect both invasion and egress are dinitroanilines (27) and diaryl ureas (28), which are suspected to destabilize microtubules and myosin tail interactions, respectively.

Inhibition of DNA synthesis.

The ability to replicate genetic information is a central aspect of reproduction for all organisms. During this process, T. gondii has a number of salvage pathways and host-scavenging properties to support its nucleotide needs. This is one process in which T. gondii differs significantly from its apicomplexan relative, Plasmodium falciparum. While P. falciparum performs almost exclusively de novo pyrimidine synthesis, T. gondii balances salvage with de novo synthesis of pyrimidine nucleotides (29). Additionally, purine de novo synthesis is not performed in T. gondii; rather, the precursors are imported from the host through specialized nucleobase transporters (29).

Because of the diversity found in eukaryotic DNA synthesis, many clinically available antiparasitics are capable of targeting this process. Pyrimethamine and sulfadiazine, two of the most important clinically used drugs in the treatment of toxoplasmosis, fall within this category. These drugs target dihydrofolate reductase (DHFR) and dihydropteroate synthase (DHPS), respectively; both are enzymes active in the folate pathway (30). Without folate, the synthesis of new thymidine nucleotides is halted, eventually interrupting the cell cycle of the parasites. Unfortunately, specific inhibition of parasite DHFR is not possible with pyrimethamine, due to its homology with human DHFR; thus, host toxicity is high. Other inhibitors of DHFR have been extensively studied and documented elsewhere in the literature (3134) and include 2,4-diaminopyrido 2,3-dipyrimidines (35), experimental sulfonamides (36), 2,4-diaminopteridines, and triazines. Based on available in vitro data, the most promising of the experimental DHFR inhibitors are the triazines; one of these, compound 14, has an IC50 of 0.02 μM (37).

Sulfadiazine demonstrates greater parasite specificity because of the suspected noneukaryotic origin of the DHPS enzyme; it is not found in humans or most other higher eukaryotes (38). A variety of novel sulfonamides, which target DHPS, have also demonstrated efficacy against T. gondii, such as compound 16, with an IC50 of 0.05 μM (36, 39) (Table 1). Other inhibitors of the DNA synthesis pathway in T. gondii generally target enzymes involved in the synthesis of individual nucleotides. Inosine-5′-monophosphate dehydrogenase, an important component of guanine synthesis, is inhibited by phthalazinones (40) through interaction at the NAD binding site (41). Additionally, parasite adenosine kinase, used in purine incorporation, binds 6-benzylthioinosine analogues as subversive substrates (42).

Most eukaryotes utilize methylation as a form of epigenetic regulation of transcription; however, in Toxoplasma tachyzoites, this has not been shown to be a source of control (43). Instead, S-adenosylmethionine (SAM)-dependent methyltransferases, which are not found in humans, are critically important in multiple parasitic metabolic pathways due to their other methylation functions (e.g., protein tagging). One compound, 1-[4-(4-nitrophenoxy)phenyl]propane-1-one (compound 21) (Table 1), is thought to affect class I SAM-dependent methyltransferases, though vacuolar ATP synthase subunit C is another potential target (44).

Inhibition of steroid synthesis.

T. gondii is capable of synthesizing some steroids that it requires, and it is dependent on the host for others (45, 46). It has been shown that the mevalonate pathway, present and functional in most eukaryotes, including humans, does not function in T. gondii, likely forcing the parasite to scavenge from the host or use alternative pathways to acquire needed steroids (47). One alternative pathway is the 1-deoxy-d-xylulose-5-phosphate (DOXP) pathway, which produces isopentenyl diphosphate and dimethylallyl diphosphate, both precursors to isoprenoids and essential molecules for parasite membrane support and cellular signaling (48, 49). The aryloxyphenoxy derivatives (Table 1) inhibit the synthesis of these precursors, preventing the formation of isoprenoids in the parasite, thereby disrupting multiple metabolic pathways and membrane structures within T. gondii. In more recent work (50), acyl coenzyme A (CoA):cholesterol transferase (ACAT) inhibitors were found to prevent cholesterol esterification by the parasite, an integral step of cholesterol uptake from the host (51) (Table 1).

Additional proposed inhibitors of T. gondii's sterol synthesis have focused on therapies that target the parasite's bifunctional protein, farnesyl diphosphate/geranylgeranyl diphosphate synthase (TgFPPS) (47). Multiple classes of TgFPPS inhibitors have been identified, including bisphosphonates and quinuclidines (Table 1). Of these, a quinuclidine, compound 27, has the lowest IC50, at 0.19 μM; however, it has not been tested in vivo (52, 53). Many bisphosphonates are able to act on T. gondii specifically; however, the host's farnesyl diphosphate synthase and geranylgeranyl diphosphate synthase are inhibited by one subclass of bisphosphonates which also inhibits TgFPPS (47). Because of this, it has been suggested that the use of statin drugs in combination with antiparasitic treatment may be beneficial. Azasteroids do not have a defined mechanism of action in T. gondii, although based on data from other parasites, it is believed to be either due to the unregulated uptake of sterols, which causes damage to organelles and membranes, or due to the potential effects on the methylation status of genes related to phospholipid biosynthesis (54). One such compound, azasteroid, compound 26, has a promising IC50 of 0.12 μM, but it is yet to be tested for in vivo activity (55) (Table 1).

Fatty acid synthesis inhibition.

Fatty acid synthesis (FAS) in apicomplexans is a process distinct from that of its analogue in mammals because of the presence of the apicoplast, an organelle acquired via secondary endosymbiosis (56). The fatty acids produced in the FASII pathway of the apicoplast are critical for organelle biogenesis and survival, despite the ability of T. gondii to scavenge some fatty acids from the host cell (57, 58). Therefore, FASII pathway enzymes are attractive drug targets. One such enzyme is β-ketoacyl-acyl carrier protein KASI/II, which plays a role in elongation of fatty acids in the FASII pathway and is specifically inhibited by thiolactomycin analogues in prokaryotes, and likely in T. gondii (57, 59, 60) (Table 1). Another FASII enzyme believed to be a drug target in fatty acid synthesis is enoyl acyl-carrier protein reductase, an enzyme responsible for the second reduction step in FASII (61). Inhibition of this enzyme's NAD+-complexed form is easily achieved with triclosan, a common antibacterial found in soaps and other personal hygiene products; however, the NADH-complexed form is not targeted by this common antibiotic (62). Therefore, various benzimidazoles, which target the reduced form of the enzyme in bacterial organisms, have a potential MoA in T. gondii; however, none of these benzimidazoles has been shown to inhibit T. gondii enoyl reductase isolated protein. Instead, in vitro analyses of the parasite's susceptibility to the compounds have revealed that the compound has other unknown parasiticidal mechanisms. The target is still thought to involve FAS due to structural similarity to a known compound, chlormidazole, which has activity against a FAS methylase in organisms that, like T. gondii, lack the FASII pathway (62).

Outside of the FASII pathway, the parasite synthesizes other fatty acids not found in humans, including pantothenate. The pantothenate pathway in T. gondii is thought to be conserved from a common prokaryotic ancestor or to have been acquired through horizontal gene transfer (63). Due to its similarity to the prokaryotic enzyme, the parasite's pantothenate synthetase can be specifically targeted with acylsulfonamides, inhibitors originally developed for Mycobacterium tuberculosis (63) (Table 1). Acylsulfonamide, compound 28, inhibits T. gondii growth in vitro with an IC50 of 0.02 μM, and it exhibits a high therapeutic index compared to other fatty acid synthesis inhibitors, with a median toxic dose (TD50) of >1 mM on HFF host cells.

Inhibition of virulence factors or host interactions.

Apicomplexan parasites manipulate the host cell's signaling pathways, transcription, cytoskeletal organization, and other cellular components in order to create an environment in which they can survive (6468). Without this ability, the host cell's innate defense mechanisms could overcome the parasite and prevent its reproduction or egress (69). Pyridinylimidazole, compound 31, (70) inhibits T. gondii in vitro with an IC50 of 0.8 to 5 μM and inhibits the activation of activin-like kinases 4, 5, and 7, which are specific host cell signaling receptors for the activation of hypoxia-inducible factor 1, a host cell transcription factor required for T. gondii growth that is manipulated by the parasite in infected host cells (71) (Table 1). In addition to potently inhibiting this pathway, compound 31 also inhibits mitogen-activated protein (MAP) kinase 1, a parasite enzyme involved in invasion (72).

Inhibition of transcription.

Transcriptional regulation is important for proper functioning of parasitic metabolism and is partially attained through chromatin packaging (73). Apicomplexan parasites and mammals use histone acetylation as a major source of regulation, and the histone deacetylation enzymes are nearly homologous (74). For this reason, histone hyperacetylation or deacetylation is not a parasite-specific process, which has been demonstrated with compound 33 (75), a fungal metabolite that simultaneously targets histone deacetylase in T. gondii and the host, as assessed by enzyme binding assays (Table 1). Despite this challenge, some investigators have identified compounds that affect histone deacetylase (HDAC) processes, such as enzyme HDAC3, in a more apicomplexan-specific manner (76). Compounds 34 and 35 are two examples of this, which have T. gondii IC50s of 7.6 nM and 39 nM, respectively. Host cell toxicity remains a concern with compound 34; the TD50 for human host cells is less than 10 times greater than the IC50 for T. gondii, but compound 35 seems to fare better on host cells with a TD50 of >10 μM in HS68 cells (74, 77) (Table 1). Garcinol, derived from the Garcinia indica fruit, is another compound that affects histone acetylation, targeting a family of lysine acetyltransferases (KAT) enzymes required for T. gondii tachyzoites to replicate successfully (78). Additionally, other (nonhistone) substrates of these compounds may be affected, contributing to the overall host cell toxicity and to the efficacy of the compound against the parasite (79). Indeed, HDACs have various roles in many biological processes, including DNA repair and cell cycle regulation.

Inhibition of reproduction or differentiation.

T. gondii has two distinct asexual life stages in humans: the tachyzoite, the stage associated with acute infection, and the bradyzoite, associated with the latent infection (80). Replication of the parasite within the parasitophorous vacuole or cyst is achieved through mitosis. Lactacystin, compound 38, is a recently discovered proteasome inhibitor capable of interfering with mitosis; parasites treated with this compound do not perish, but rather halt cellular replication until the compound has been metabolized or removed, even if the treatment period lasts for several days (81). Because of this, no IC50 could be generated for this class of inhibitor, although significant effects were noted at 2 μM. While the parasitostatic effects of compound 38 present limitations to development, no host cell toxicity was noted even at the highest compound concentration studied (2 μM).

When the parasite encounters stressful conditions, such as an immune system response or metabolic insult, the rapidly dividing tachyzoite undergoes differentiation to the slowly replicating bradyzoite (82). This shift significantly changes the genetic expression profile of T. gondii and leads to a slower-growing, hardier encysted form of the parasite that localizes to the brain and muscular tissue of the host. Two lactate dehydrogenase (LDH) isoforms are differentially expressed during the tachyzoite and bradyzoite stages. LDH1 is expressed mainly in the tachyzoite stage, while LDH2 is expressed during the bradyzoite stage; both are substantially different from the mammalian LDH (83). The cotton plant-derived gossypol and its derivatives (exemplified by compound 37) have been shown to be potent LDH2 inhibitors (enzyme Ki, 1.1 μM) and are somewhat active against LDH1 (enzyme Ki, 6.1 μM), with an IC50 of 5 to 10 μM against intracellular tachyzoites (84). However, these compounds have not been tested against the bradyzoite form.

Effects on reactive oxygen species regulation.

Through the combination of the CD8+ T-cell response and interferon gamma-mediated activation, a competent host immune system is capable of subduing most of the tachyzoites produced in an acute T. gondii infection. One facet of this response is the production of reactive oxygen species (ROS) by macrophages, a response induced by cationic dyes (85), quinoline derivatives (86), and various alkaloids (87) (Table 1). Alkaloid compounds 44 and 45 have the lowest IC50 values in this category (0.7 nM), but host cell toxicity is a concern: in THP-1 host cells, compound 44 demonstrated a TD50 of 1 nM, while compound 45 had a more favorable TD50 of 0.392 μM (86).

Quinones, though extensively studied, do not have an established MoA in Toxoplasma. A parasite-specific NADH dehydrogenase type II complex, essential to cellular respiration, binds some quinones with high affinity, indicating that this may be one target of this compound class (88). Without the ability to reduce oxygen into water, the production of ROS would be promoted, likely causing damage to parasite mitochondria (89).

COMPOUNDS WITHOUT KNOWN OR SUSPECTED MODES OF ACTION

Compounds that lack a known or suspected MoA, but demonstrate anti-Toxoplasma activity, present an opportunity for further exploration. Of note, pyrimethamine, atovaquone, and sulfadiazine did not have a known MoA at the time that they were FDA approved; thus, the lack of a MoA does not necessarily prevent an otherwise-promising compound from progressing in development.

Of the compounds that do not have specific mechanisms elucidated, one stands out as particularly interesting: a semisynthetic artemisinin, compound 54. Artemisinin, the precursor of all of the semisynthetic artemisinin derivatives used to treat malaria, was isolated from the plant Artemisia annua (90). Compound 54 has poor aqueous solubility (91); however, due to its high lipophilicity, compound 54 is suspected to have high blood-brain barrier penetration; indeed, further studies on reactivation of latent T. gondii infections have shown that this compound has some ability to protect mice from immunosuppression-induced reactivated infections (90). Because of the potency of this compound, as well as closely related derivatives (artemisone) passing FDA phase I trials (92), compound 54 and other artemisinin derivatives are promising candidates for further study.

Other compound groups with no known MoA include metals and metal complexes; images taken from one silver exposure study demonstrated significant morphological alterations, indicating that the MoA likely involves some form of mechanical damage to the cellular surface of the parasite (Table 2). Of these compounds, the highly effective metal ion complex, compound 50 (IC50, 0.0187 μM) appears to be the most the most promising (93).

COMPOUNDS IDENTIFIED IN SCREENS

Most clinically available anti-Toxoplasma drugs were originally developed to treat infections caused by other protozoal or bacterial pathogens (94). Traditionally, compounds active against Plasmodium were often subsequently tested against T. gondii. More recently, T. gondii-specific high-throughput screens (HTS) are becoming more common for discovery of compounds active against this protozoan. However, the downside of screening is that little information other than the compound structure and IC50 data are readily gathered, leaving significant work to detail additional features. For the purpose of this review, T. gondii chemical screens were classified into 4 different groups: (i) cell-based phenotypic screens; (ii) target-based in vitro biochemical screens; (iii) cheminformatics-based virtual screens; (iv) drug-repurposing screens. The inherent value of such screens, as seen in Table 3, is to provide starting points for subsequent characterization and optimization.

Cell-based phenotypic screens.

In cell-based phenotypic screens, compounds are evaluated against live parasites by measuring the in vitro parasite replication rate by using strains that express β-galactosidase or fluorescent proteins, or by employing the [3H]uracil incorporation assay (9597). As an example of the power of this approach, Table 3 presents a list of the promising compounds discovered using cell-based screens. For example, a small focused screen (98) identified 8 compounds with a biphenylimidazoazine scaffold with IC50s of ≤0.6 μM against T. gondii (Table 3); these compounds also demonstrated broad antiparasitic effects against several apicomplexan parasites, including P. falciparum, Neospora caninum, Eimeria tenella, and Besnoitia besnoiti, suggesting that they were rather nonspecific inhibitors of T. gondii. In a recent high-throughput screnning (HTS) approach (99), 6,811 compounds were screened, and N-4-ethyl-benzoyl-2-hydroxybenzamide, compound 80, was identified, with an IC90 value of ∼0.031 μM. Using an insertional mutagenesis approach, the authors identified a putative target gene, adaptin-3β, which is part of a secretory protein complex responsible for the secretion of micronemes, rhoptries, and dense granules. This compound was optimized to yield compound 81, which has an IC90 value of ∼0.016 μM. Compounds 80 and 81, at respective doses of 50 and 20 mg/kg/day, protected mice against T. gondii acute infection. However, these compounds had low specificity for T. gondii, as they were also active against other protozoan parasites, including Trypanosoma brucei rhodesiense, Trypanosoma cruzi, Leishmania donovani, and P. falciparum (100). Another phenotypic study identified endochin-like quinolone compounds 68 and 69 with in vitro IC50s of 0.1 and 0.007 nM, respectively (101) (Table 3). These compounds are orally bioavailable and, at doses of 0.3 to 1.0 mg/kg/day given once a day for 5 days, prevented acute toxoplasmosis in a murine experimental model. Moreover, these compounds drastically reduced the cyst burden in a latent murine infection model. Their low IC50 combined with the ability to inhibit acute and chronic forms of infection makes these compounds promising anti-Toxoplasma therapeutic leads.

In addition to measuring in vitro growth rate, some T. gondii phenotypic screens identified compounds that inhibited host cell attachment and invasion. Using this approach, a focused small-molecule screen identified cysteine protease inhibitors that block invasion (102). Other screens have used dual-fluorescent image-based high-content screening (HCS) to directly observe parasite attachment and entry into the host cells (103, 104). Even though phenotypic screens are capable of identifying new chemical entities, one drawback of the phenotype-based approach is the difficulty in identifying target proteins. To circumvent this problem, Hall et al. used a combination of HCS and chemical genetics to screen a library of 1,222 inhibitors that covalently modified the target protein and then applied click chemistry along with tandem orthogonal proteolysis activity-based protein profiling to identify target proteins. With this approach, they identified compound 67, which blocks host cell invasion by inhibiting the activity of TgDJ-1, a unique regulator of microneme secretion and motility (104). These studies suggest that phenotypic screens combined with chemical genetics or forward genetic screens (69), such as drug-resistant strain generation via insertional mutagenesis or chemical mutagenesis, are powerful approaches to identify new drugs and drug targets.

Target-based in vitro biochemical screens.

Target-based in vitro biochemical screens usually involve a biochemical assay with a purified recombinant protein. One of the major advantages of a target-based screen is the possibility of screening or designing drugs based on the enzymatic property or the crystal structure of a protein. These screens were carried out on T. gondii even before the availability of its genome sequence (105). Most of these screens were focused on developing inhibitors against known essential enzymes, such as DHFR, DHPS, and nucleoside triphosphate hydrolase (NTPase). However, due to the toxicity of clinically available antifolate drugs, many of the initial screens were focused on identifying additional compounds that specifically inhibited T. gondii DHFR (33, 34, 106111) and DHPS (39, 112) while avoiding host effects. Most of these small-scale screens were performed to identify compounds that were active against these enzymes, but very few of these studies report T. gondii in vivo data (37, 113, 114). One of the early target-based HTS in T. gondii was performed by Asai et al., who screened 150,000 compounds against Tg-NTPases, which are essential proteins for T. gondii tachyzoite replication inside the parasitophorous vacuole (115) (Table 3). In this screen, compounds 89, 90, and 91 were identified that inhibited T. gondii growth in vitro with IC50s of ≤7.0 μM and inhibited NTPase I and II enzymes with IC50s ranging from 1.3 μM to 14.5 μM.

Several target-based drug screens were also performed against T. gondii CDPKs because, as described above, these kinases regulate essential biological processes, such as cell division, development, motility, microneme secretion, invasion, and egress (116). In addition, the ATP binding pockets of most of the apicomplexan CDPKs have a gatekeeper residue with a smaller side chain, whereas most of the ATP binding pockets in mammalian serine-threonine protein kinases have a gatekeeper amino acid with a bulky side chain. This property has been exploited to develop inhibitors specific for TgCDPK1, an essential kinase that regulates gliding motility, invasion, and egress in T. gondii (117). Based on structure-activity relationship (SAR) studies, a number of pyrazolopyrimidines were identified which specifically inhibit TgCDPK1 and prevent T. gondii growth at concentrations below 1 μM (24, 118, 119). One of these, compound 86, is orally bioavailable and can reach tachyzoites in the brains of infected mice by crossing the blood-brain barrier and reach therapeutic concentrations (120). Interestingly, compound 86 has broad-spectrum antiparasitic activity against several pathogens, such as Cryptosporidium parvum (121), Neospora caninum (122), and Plasmodium falciparum (123).

Cheminformatics-based virtual screens.

Cheminformatics-based virtual screens are being performed to identify novel anti-Toxoplasma compounds (87, 124, 125). The availability of crystal structures for some of the T. gondii proteins and the improvement in computational molecular modeling approaches facilitate this approach. For example, a chemoinformatic virtual screen against rhoptry protein 18 (ROP18) led to the identification of compound 93, which inhibits T. gondii growth in vitro with an IC50 of ∼2 nM (Table 3). In this screen, the authors performed in silico chemical searches on 45,384 commercially available compounds and identified 17 of these, based on their expected activity and ADME characteristics. From these 17 compounds, those authors identified compound 86 to be most potent against T. gondii in vitro. Even though compound 93 was identified as a ROP18 inhibitor, this compound has additional unidentified effects on the parasite.

Drug-repurposing screens.

Drug-repurposing screens are common in T. gondii drug discovery because drugs active against one apicomplexan parasite are often active against other apicomplexans, due to the conservation of biochemical pathways between these parasites. As a result, compounds active against Plasmodium falciparum are often screened against T. gondii. Many compounds now in clinical use are a result of these investigations (13). Screening of the open-access Medicines for Malaria Venture (MMV) Malaria Box led to the identification of 7 anti-Toxoplasma compounds with novel chemical scaffolds (126) (Table 3), the most potent of which was compound 94, with an IC50 of 0.19 μM (126). In another screen, approximately 15 broad-spectrum anti-Toxoplasma compounds, with IC50s below 10 μM, were identified by screening a library of 309,474 unique compounds against P. falciparum and testing a set of representative compounds against T. gondii, Leishmania major, and Trypanosoma brucei.

CONCLUSION

Within this review, more than 50 chemotypes have been described. Based on the identified characteristics, a number of specific compounds stand out as particularly notable. Pyridinylpyrrole, compound 3, demonstrated 90% increased survival and low organ parasite loads during an in vivo trial, with no sign of recrudescence 12 months after the completion of treatment (19, 21) (Table 1). Another promising compound class which inhibits fatty acid synthesis are the acylsulfonamides, the molecular target for which is not present in the host cell (63) (Table 1). Compound 28, an acylsulfonamide, does not have any reported in vivo data, but it has very low host cell toxicity in HFF cells (TD50, >1 mM) compared to its T. gondii IC50 (0.02 μM), indicating it may have a relatively high therapeutic index. One of the most promising compounds identified via a screening assay was compound 86, a semisynthetic artemisinin (120). This compound is orally bioavailable and capable of crossing the blood-brain barrier, with a survival increase of 60% in treated mice (90). The endochin-like quinolones (compounds 68 and 69), also identified via screening, additionally demonstrate promising characteristics, such as a high therapeutic index and efficacy against the acute and chronic stages in vitro (Table 3).

Inhibition of translation is common against prokaryotic targets, but due to homology to host ribosomal components, it is rarely utilized in antiparasitic modes of action. Future work that targets mechanisms of translational control may offer additional avenues for therapy (128).

In summary, with the expanding number of immunocompromised individuals at high risk, the demand for new toxoplasmosis treatment options is rising. Some of the novel compounds reviewed here may represent good starting points for the discovery of effective new drugs against T. gondii. Increasing the medicinal arsenal of anti-Toxoplasma therapeutics may yield a potent and less toxic option for future patients.

Supplementary Material

Supplemental material

Funding Statement

This work was funded by HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID AI116723) (J.L.V.) and HHS| NIH | National Institute of General Medical Sciences (NIGMS GM103427) (P.H.D.). Additionally, the following support is acknowledged: the Nebraska Research Initiative (P.H.D.) and the University of Nebraska at Omaha FUSE and GRACA (M.M.M.).

Footnotes

Supplemental material for this article may be found at http://dx.doi.org/10.1128/AAC.01176-16.

REFERENCES

  • 1.Dubey JP. 2008. The history of Toxoplasma gondii: the first 100 years. J Eukaryot Microbiol 55:467–475. doi: 10.1111/j.1550-7408.2008.00345.x. [DOI] [PubMed] [Google Scholar]
  • 2.Rengifo-Herrera C, Ortega-Mora LM, Alvarez-García G, Gómez-Bautista M, García-Párraga D, García-Peña FJ, Pedraza-Díaz S. 2012. Detection of Toxoplasma gondii antibodies in Antarctic pinnipeds. Vet Parasitol 190:259–262. doi: 10.1016/j.vetpar.2012.05.020. [DOI] [PubMed] [Google Scholar]
  • 3.Dhama K, Rajagunalan S, Chakraborty S, Verma AK, Kumar A, Tiwari R, Kapoor S. 2013. Food-borne pathogens of animal origin-diagnosis, prevention, control and their zoonotic significance: a review. Pak J Biol Sci 16:1076–1085. doi: 10.3923/pjbs.2013.1076.1085. [DOI] [PubMed] [Google Scholar]
  • 4.Swierzy IJ, Muhammad M, Kroll J, Abelmann A, Tenter AM, Lüder CGK. 2014. Toxoplasma gondii within skeletal muscle cells: a critical interplay for food-borne parasite transmission. Int J Parasitol 44:91–98. doi: 10.1016/j.ijpara.2013.10.001. [DOI] [PubMed] [Google Scholar]
  • 5.Pittman KJ, Knoll LJ. 2015. Long-term relationships: the complicated interplay between the host and the developmental stages of Toxoplasma gondii during acute and chronic infections. Microbiol Mol Biol Rev 79:387–401. doi: 10.1128/MMBR.00027-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Meireles LR, Ekman CC, Andrade HF Jr, Luna EJ. 2015. Human toxoplasmosis outbreaks and the agent infecting form. Findings from a systematic review. Rev Inst Med Trop São Paulo 57:369–376. doi: 10.1590/S0036-46652015000500001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Peters PJ, Thigpen MC, Parise ME, Newman RD. 2007. Safety and toxicity of sulfadoxine/pyrimethamine: implications for malaria prevention in pregnancy using intermittent preventive treatment. Drug Saf 30:481–501. doi: 10.2165/00002018-200730060-00003. [DOI] [PubMed] [Google Scholar]
  • 8.Pissinate K, dos Santos Martins-Duarte É, Schaffazick SR, de Oliveira CP, Vommaro RC, Guterres SS, Pohlmann AR, de Souza W. 2014. Pyrimethamine-loaded lipid-core nanocapsules to improve drug efficacy for the treatment of toxoplasmosis. Parasitol Res 113:555–564. doi: 10.1007/s00436-013-3715-6. [DOI] [PubMed] [Google Scholar]
  • 9.Macy E, Poon K-YT. 2009. Self-reported antibiotic allergy incidence and prevalence: age and sex effects. Am J Med 122:778.e1–7. doi: 10.1016/j.amjmed.2009.01.034. [DOI] [PubMed] [Google Scholar]
  • 10.Wulf NR, Matuszewski KA. 2013. Sulfonamide cross-reactivity: is there evidence to support broad cross-allergenicity? Am J Health Syst Pharm 70:1483–1494. doi: 10.2146/ajhp120291. [DOI] [PubMed] [Google Scholar]
  • 11.McAuley JB. 2014. Congenital toxoplasmosis. J Pediatr Infect Dis Soc 3(Suppl 1):S30–S35. doi: 10.1093/jpids/piu077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Neu N, Duchon J, Zachariah P. 2015. TORCH infections. Clin Perinatol 42:77–103. doi: 10.1016/j.clp.2014.11.001. [DOI] [PubMed] [Google Scholar]
  • 13.Neville AJ, Zach SJ, Wang X, Larson JJ, Judge AK, Davis LA, Vennerstrom JL, Davis PH. 2015. Clinically available medicines demonstrating anti-Toxoplasma activity. Antimicrob Agents Chemother 59:7161–7169. doi: 10.1128/AAC.02009-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Goodwin DG, Strobl JS, Lindsay DS. 2011. Evaluation of five antischizophrenic agents against Toxoplasma gondii in human cell cultures. J Parasitol 97:148–151. doi: 10.1645/GE-2536.1. [DOI] [PubMed] [Google Scholar]
  • 15.Sepulveda-Arias JC, Veloza LA, Mantilla-Muriel LE. 2014. Anti-Toxoplasma activity of natural products: a review. Recent Pat Antiinfect Drug Discov 9:186–194. doi: 10.2174/1574891X1066615041010321. [DOI] [PubMed] [Google Scholar]
  • 16.Kamerkar S, Davis PH. 2012. Toxoplasma on the brain: understanding host-pathogen interactions in chronic CNS infection. J Parasitol Res 2012:589295. doi: 10.1155/2012/589295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhou W, Wang Y, Lu A, Zhang G. 2016. Systems pharmacology in small molecular drug discovery. Int J Mol Sci 17:246. doi: 10.3390/ijms17020246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Roiko MS, Carruthers VB. 2009. New roles for perforins and proteases in apicomplexan egress. Cell Microbiol 11:1444–1452. doi: 10.1111/j.1462-5822.2009.01357.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nare B, Allocco JJ, Liberator PA, Donald RGK. 2002. Evaluation of a cyclic GMP-dependent protein kinase inhibitor in treatment of murine toxoplasmosis: gamma interferon is required for efficacy. Antimicrob Agents Chemother 46:300–307. doi: 10.1128/AAC.46.2.300-307.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhang C, Ondeyka JG, Herath KB, Guan Z, Collado J, Platas G, Pelaez F, Leavitt PS, Gurnett A, Nare B, Liberator P, Singh SB. 2005. Tenellones A and B from a Diaporthe sp.: two highly substituted benzophenone inhibitors of parasite cGMP-dependent protein kinase activity. J Nat Prod 68:611–613. doi: 10.1021/np049591n. [DOI] [PubMed] [Google Scholar]
  • 21.Wiersma HI, Galuska SE, Tomley FM, Sibley LD, Liberator PA, Donald RGK. 2004. A role for coccidian cGMP-dependent protein kinase in motility and invasion. Int J Parasitol 34:369–380. doi: 10.1016/j.ijpara.2003.11.019. [DOI] [PubMed] [Google Scholar]
  • 22.Donald RGK, Allocco J, Singh SB, Nare B, Salowe SP, Wiltsie J, Liberator PA. 2002. Toxoplasma gondii cyclic GMP-dependent kinase: chemotherapeutic targeting of an essential parasite protein kinase. Eukaryot Cell 1:317–328. doi: 10.1128/EC.1.3.317-328.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lavine MD, Arrizabalaga G. 2007. Invasion and egress by the obligate intracellular parasite Toxoplasma gondii: potential targets for the development of new antiparasitic drugs. Curr Pharm Des 13:641–651. doi: 10.2174/138161207780162854. [DOI] [PubMed] [Google Scholar]
  • 24.Johnson SM, Murphy RC, Geiger JA, DeRocher AE, Zhang Z, Ojo KK, Larson ET, Perera BGK, Dale EJ, He P, Reid MC, Fox AMW, Mueller NR, Merritt EA, Fan E, Parsons M, Van Voorhis WC, Maly DJ. 2012. Development of Toxoplasma gondii calcium-dependent protein kinase 1 (TgCDPK1) inhibitors with potent anti-Toxoplasma activity. J Med Chem 55:2416–2426. doi: 10.1021/jm201713h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhang Z, Ojo KK, Johnson SM, Larson ET, He P, Geiger JA, Castellanos-Gonzalez A, White AC, Parsons M, Merritt EA, Maly DJ, Verlinde CLMJ, Van Voorhis WC, Fan E. 2012. Benzoylbenzimidazole-based selective inhibitors targeting Cryptosporidium parvum and Toxoplasma gondii calcium-dependent protein kinase-1. Bioorg Med Chem Lett 22:5264–5267. doi: 10.1016/j.bmcl.2012.06.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lourido S, Tang K, Sibley LD. 2012. Distinct signalling pathways control Toxoplasma egress and host-cell invasion. EMBO J 31:4524–4534. doi: 10.1038/emboj.2012.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Endeshaw MM, Li C, de Leon J, Yao N, Latibeaudiere K, Premalatha K, Morrissette N, Werbovetz KA. 2010. Synthesis and evaluation of oryzalin analogs against Toxoplasma gondii. Bioorg Med Chem Lett 20:5179–5183. doi: 10.1016/j.bmcl.2010.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kortagere S, Mui E, McLeod R, Welsh WJ. 2011. Rapid discovery of inhibitors of Toxoplasma gondii using hybrid structure-based computational approach. J Comput Aided Mol Des 25:403–411. doi: 10.1007/s10822-011-9420-6. [DOI] [PubMed] [Google Scholar]
  • 29.Hyde JE. 2007. Targeting purine and pyrimidine metabolism in human apicomplexan parasites. Curr Drug Targets 8:31–47. doi: 10.2174/138945007779315524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tessema SK, Kassa M, Kebede A, Mohammed H, Leta GT, Woyessa A, Guma GT, Petros B. 2015. Declining trend of Plasmodium falciparum dihydrofolate reductase (dhfr) and dihydropteroate synthase (dhps) mutant alleles after the withdrawal of sulfadoxine-pyrimethamine in North Western Ethiopia. PLoS One 10:e0126943. doi: 10.1371/journal.pone.0126943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Anderson AC. 2005. Targeting DHFR in parasitic protozoa. Drug Discov Today 10:121–128. doi: 10.1016/S1359-6446(04)03308-2. [DOI] [PubMed] [Google Scholar]
  • 32.Tawari NR, Bag S, Degani MS. 2011. A review of molecular modelling studies of dihydrofolate reductase inhibitors against opportunistic microorganisms and comprehensive evaluation of new models. Curr Pharm Des 17:712–751. doi: 10.2174/138161211795428966. [DOI] [PubMed] [Google Scholar]
  • 33.Bag S, Tawari NR, Degani MS, Queener SF. 2010. Design, synthesis, biological evaluation and computational investigation of novel inhibitors of dihydrofolate reductase of opportunistic pathogens. Bioorg Med Chem 18:3187–3197. doi: 10.1016/j.bmc.2010.03.031. [DOI] [PubMed] [Google Scholar]
  • 34.Zaware N, Sharma H, Yang J, Devambatla RKV, Queener SF, Anderson KS, Gangjee A. 2013. Discovery of potent and selective inhibitors of Toxoplasma gondii thymidylate synthase for opportunistic infections. ACS Med Chem Lett 4:1148–1151. doi: 10.1021/ml400208v. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rosowsky A, Cody V, Galitsky N, Fu H, Papoulis AT, Queener SF. 1999. Structure-based design of selective inhibitors of dihydrofolate reductase: synthesis and antiparasitic activity of 2,4-diaminopteridine analogues with a bridged diarylamine side chain. J Med Chem 42:4853–4860. doi: 10.1021/jm990331q. [DOI] [PubMed] [Google Scholar]
  • 36.Doliwa C, Escotte-Binet S, Aubert D, Sauvage V, Velard F, Schmid A, Villena I. 2013. Sulfadiazine resistance in Toxoplasma gondii: no involvement of overexpression or polymorphisms in genes of therapeutic targets and ABC transporters. Parasite 20:19. doi: 10.1051/parasite/2013020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mui EJ, Schiehser GA, Milhous WK, Hsu H, Roberts CW, Kirisits M, Muench S, Rice D, Dubey JP, Fowble JW, Rathod PK, Queener SF, Liu SR, Jacobus DP, McLeod R. 2008. Novel triazine JPC-2067-B inhibits Toxoplasma gondii in vitro and in vivo. PLoS Negl Trop Dis 2:e190. doi: 10.1371/journal.pntd.0000190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hammoudeh DI, Daté M, Yun M-K, Zhang W, Boyd VA, Viacava Follis A, Griffith E, Lee RE, Bashford D, White SW. 2014. Identification and characterization of an allosteric inhibitory site on dihydropteroate synthase. ACS Chem Biol 9:1294–1302. doi: 10.1021/cb500038g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chio LC, Bolyard LA, Nasr M, Queener SF. 1996. Identification of a class of sulfonamides highly active against dihydropteroate synthase form Toxoplasma gondii, Pneumocystis carinii, and Mycobacterium avium. Antimicrob Agents Chemother 40:727–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hedstrom L. 2009. IMP dehydrogenase: structure, mechanism and inhibition. Chem Rev 109:2903–2928. doi: 10.1021/cr900021w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Johnson CR, Gorla SK, Kavitha M, Zhang M, Liu X, Striepen B, Mead JR, Cuny GD, Hedstrom L. 2013. Phthalazinone inhibitors of inosine-5′-monophosphate dehydrogenase from Cryptosporidium parvum. Bioorg Med Chem Lett 23:1004–1007. doi: 10.1016/j.bmcl.2012.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kim YA, Rawal RK, Yoo J, Sharon A, Jha AK, Chu CK, Rais RH, Al Safarjalani ON, Naguib FNM, El Kouni MH. 2010. Structure-activity relationships of carbocyclic 6-benzylthioinosine analogues as subversive substrates of Toxoplasma gondii adenosine kinase. Bioorg Med Chem 18:3403–3412. doi: 10.1016/j.bmc.2010.04.003. [DOI] [PubMed] [Google Scholar]
  • 43.Gissot M, Choi S-W, Thompson RF, Greally JM, Kim K. 2008. Toxoplasma gondii and Cryptosporidium parvum lack detectable DNA cytosine methylation. Eukaryot Cell 7:537–540. doi: 10.1128/EC.00448-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Choi H-J, Lee J-H, Yeo S-J, Kaewintajuk K, Yi K-Y, Kim S, Song H-O, Park H. 2015. Evaluation of anti-coccidial effects of 1-[4-(4-nitrophenoxy)phenyl] propane-1-one and identification of its potential target proteins in Toxoplasma gondii. Arch Pharm Res 38:752–760. doi: 10.1007/s12272-014-0400-y. [DOI] [PubMed] [Google Scholar]
  • 45.Coppens I, Sinai AP, Joiner KA. 2000. Toxoplasma gondii exploits host low-density lipoprotein receptor-mediated endocytosis for cholesterol acquisition. J Cell Biol 149:167–180. doi: 10.1083/jcb.149.1.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Charron AJ, Sibley LD. 2002. Host cells: mobilizable lipid resources for the intracellular parasite Toxoplasma gondii. J Cell Sci 115:3049–3059. [DOI] [PubMed] [Google Scholar]
  • 47.Li Z-H, Ramakrishnan S, Striepen B, Moreno SNJ. 2013. Toxoplasma gondii relies on both host and parasite isoprenoids and can be rendered sensitive to atorvastatin. PLoS Pathog 9:e1003665. doi: 10.1371/journal.ppat.1003665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Elicio PD, Chao MN, Galizzi M, Li C, Szajnman SH, Docampo R, Moreno SNJ, Rodriguez JB. 2013. Design, synthesis and biological evaluation of WC-9 analogs as antiparasitic agents. Eur J Med Chem 69:480–489. doi: 10.1016/j.ejmech.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Guggisberg AM, Amthor RE, Odom AR. 2014. Isoprenoid biosynthesis in Plasmodium falciparum. Eukaryot Cell 13:1348–1359. doi: 10.1128/EC.00160-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wanke M, Skorupinska-Tudek K, Swiezewska E. 2001. Isoprenoid biosynthesis via 1-deoxy-d-xylulose 5-phosphate/2-C-methyl-d-erythritol 4-phosphate (DOXP/MEP) pathway. Acta Biochim Pol 48:663–672. [PubMed] [Google Scholar]
  • 51.Sonda S, Ting LM, Novak S, Kim K, Maher JJ, Farese RV, Ernst JD. 2001. Cholesterol esterification by host and parasite is essential for optimal proliferation of Toxoplasma gondii. J Biol Chem 276:34434–34440. doi: 10.1074/jbc.M105025200. [DOI] [PubMed] [Google Scholar]
  • 52.Martins-Duarte ES, Urbina JA, de Souza W, Vommaro RC. 2006. Antiproliferative activities of two novel quinuclidine inhibitors against Toxoplasma gondii tachyzoites in vitro. J Antimicrob Chemother 58:59–65. doi: 10.1093/jac/dkl180. [DOI] [PubMed] [Google Scholar]
  • 53.Ferrer-Casal M, Li C, Galizzi M, Stortz CA, Szajnman SH, Docampo R, Moreno SNJ, Rodriguez JB. 2014. New insights into molecular recognition of 1,1-bisphosphonic acids by farnesyl diphosphate synthase. Bioorg Med Chem 22:398–405. doi: 10.1016/j.bmc.2013.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Martins-Duarte ES, Lemgruber L, Lorente SO, Gros L, Magaraci F, Gilbert IH, de Souza W, Vommaro RC. 2011. Evaluation of three novel azasterols against Toxoplasma gondii. Vet Parasitol 177:157–161. doi: 10.1016/j.vetpar.2010.11.034. [DOI] [PubMed] [Google Scholar]
  • 55.Dantas-Leite L, Urbina JA, de Souza W, Vommaro RC. 2004. Selective anti-Toxoplasma gondii activities of azasterols. Int J Antimicrob Agents 23:620–626. doi: 10.1016/j.ijantimicag.2003.11.005. [DOI] [PubMed] [Google Scholar]
  • 56.Fichera ME, Roos DS. 1997. A plastid organelle as a drug target in apicomplexan parasites. Nature 390:407–409. doi: 10.1038/37132. [DOI] [PubMed] [Google Scholar]
  • 57.Mazumdar J, Wilson HE, Masek K, Hunter AC, Striepen B. 2006. Apicoplast fatty acid synthesis is essential for organelle biogenesis and parasite survival in Toxoplasma gondii. Proc Natl Acad Sci U S A 103:13192–13197. doi: 10.1073/pnas.0603391103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ramakrishnan S, Docampo MD, MacRae JI, Pujol FM, Brooks CF, van Dooren GG, Hiltunen JK, Kastaniotis AJ, McConville MJ, Striepen B. 2012. Apicoplast and endoplasmic reticulum cooperate in fatty acid biosynthesis in apicomplexan parasite Toxoplasma gondii. J Biol Chem 287:4957–4971. doi: 10.1074/jbc.M111.310144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Olsen JG, Kadziola A, von Wettstein-Knowles P, Siggaard-Andersen M, Larsen S. 2001. Structures of beta-ketoacyl-acyl carrier protein synthase I complexed with fatty acids elucidate its catalytic machinery. Structure (London) 9:233–243. doi: 10.1016/S0969-2126(01)00583-4. [DOI] [PubMed] [Google Scholar]
  • 60.Martins-Duarte ES, Jones SM, Gilbert IH, Atella GC, de Souza W, Vommaro RC. 2009. Thiolactomycin analogues as potential anti-Toxoplasma gondii agents. Parasitol Int 58:411–415. doi: 10.1016/j.parint.2009.08.004. [DOI] [PubMed] [Google Scholar]
  • 61.Ramakrishnan S, Docampo MD, MacRae JI, Ralton JE, Rupasinghe T, McConville MJ, Striepen B. 2015. The intracellular parasite Toxoplasma gondii depends on the synthesis of long-chain and very long-chain unsaturated fatty acids not supplied by the host cell. Mol Microbiol 97:64–76. doi: 10.1111/mmi.13010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wilkinson C, McPhillie MJ, Zhou Y, Woods S, Afanador GA, Rawson S, Khaliq F, Prigge ST, Roberts CW, Rice DW, McLeod R, Fishwick CW, Muench SP. 2014. The benzimidazole based drugs show good activity against T. gondii but poor activity against its proposed enoyl reductase enzyme target. Bioorg Med Chem Lett 24:911–916. doi: 10.1016/j.bmcl.2013.12.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mageed SN, Cunningham F, Hung AW, Silvestre HL, Wen S, Blundell TL, Abell C, McConkey GA. 2014. Pantothenic acid biosynthesis in the parasite Toxoplasma gondii: a target for chemotherapy. Antimicrob Agents Chemother 58:6345–6353. doi: 10.1128/AAC.02640-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Franco M, Panas MW, Marino ND, Lee M-CW, Buchholz KR, Kelly FD, Bednarski JJ, Sleckman BP, Pourmand N, Boothroyd JC. 2016. A novel secreted protein, MYR1, is central to Toxoplasma's manipulation of host cells. mBio 7(1):e02231-15. doi: 10.1128/mBio.02231-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Etheridge RD, Alaganan A, Tang K, Lou HJ, Turk BE, Sibley LD. 2014. The Toxoplasma pseudokinase ROP5 forms complexes with ROP18 and ROP17 kinases that synergize to control acute virulence in mice. Cell Host Microbe 15:537–550. doi: 10.1016/j.chom.2014.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Jones NG, Wang Q, Sibley LD. 23 July 2016. Secreted protein kinases regulate cyst burden during chronic toxoplasmosis. Cell Microbiol doi: 10.1111/cmi.12651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lorenzi H, Khan A, Behnke MS, Namasivayam S, Swapna LS, Hadjithomas M, Karamycheva S, Pinney D, Brunk BP, Ajioka JW, Ajzenberg D, Boothroyd JC, Boyle JP, Dardé ML, Diaz-Miranda MA, Dubey JP, Fritz HM, Gennari SM, Gregory BD, Kim K, Saeij JPJ, Su C, White MW, Zhu X-Q, Howe DK, Rosenthal BM, Grigg ME, Parkinson J, Liu L, Kissinger JC, Roos DS, Sibley LD. 2016. Local admixture of amplified and diversified secreted pathogenesis determinants shapes mosaic Toxoplasma gondii genomes. Nat Commun 7:10147. doi: 10.1038/ncomms10147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bradley PJ, Sibley LD. 2007. Rhoptries: an arsenal of secreted virulence factors. Curr Opin Microbiol 10:582–587. doi: 10.1016/j.mib.2007.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brown KM, Suvorova E, Farrell A, McLain A, Dittmar A, Wiley GB, Marth G, Gaffney PM, Gubbels MJ, White M, Blader IJ. 2014. Forward genetic screening identifies a small molecule that blocks Toxoplasma gondii growth by inhibiting both host- and parasite-encoded kinases. PLoS Pathog 10:e1004180. doi: 10.1371/journal.ppat.1004180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wei S, Daniel BJ, Brumlik MJ, Burow ME, Zou W, Khan IA, Wadsworth S, Siekierka J, Curiel TJ. 2007. Drugs designed to inhibit human p38 mitogen-activated protein kinase activation treat Toxoplasma gondii and Encephalitozoon cuniculi infection. Antimicrob Agents Chemother 51:4324–4328. doi: 10.1128/AAC.00680-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Spear W, Chan D, Coppens I, Johnson RS, Giaccia A, Blader IJ. 2006. The host cell transcription factor hypoxia-inducible factor 1 is required for Toxoplasma gondii growth and survival at physiological oxygen levels. Cell Microbiol 8:339–352. doi: 10.1111/j.1462-5822.2005.00628.x. [DOI] [PubMed] [Google Scholar]
  • 72.Robert-Gangneux F, Creuzet C, Dupouy-Camet J, Roisin MP. 2000. Involvement of the mitogen-activated protein (MAP) kinase signalling pathway in host cell invasion by Toxoplasma gondii. Parasite (Paris) 7:95–101. doi: 10.1051/parasite/2000072095. [DOI] [PubMed] [Google Scholar]
  • 73.Sterner DE, Berger SL. 2000. Acetylation of histones and transcription-related factors. Microbiol Mol Biol Rev 64:435–459. doi: 10.1128/MMBR.64.2.435-459.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bougdour A, Maubon D, Baldacci P, Ortet P, Bastien O, Bouillon A, Barale J-C, Pelloux H, Ménard R, Hakimi M-A. 2009. Drug inhibition of HDAC3 and epigenetic control of differentiation in Apicomplexa parasites. J Exp Med 206:953–966. doi: 10.1084/jem.20082826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Darkin-Rattray SJ, Gurnett AM, Myers RW, Dulski PM, Crumley TM, Allocco JJ, Cannova C, Meinke PT, Colletti SL, Bednarek MA, Singh SB, Goetz MA, Dombrowski AW, Polishook JD, Schmatz DM. 1996. Apicidin: a novel antiprotozoal agent that inhibits parasite histone deacetylase. Proc Natl Acad Sci U S A 93:13143–13147. doi: 10.1073/pnas.93.23.13143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Vanagas L, Jeffers V, Bogado SS, Dalmasso MC, Sullivan WJ, Angel SO. 2012. Toxoplasma histone acetylation remodelers as novel drug targets. Expert Rev Anti Infect Ther 10:1189–1201. doi: 10.1586/eri.12.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Strobl JS, Cassell M, Mitchell SM, Reilly CM, Lindsay DS. 2007. Scriptaid and suberoylanilide hydroxamic acid are histone deacetylase inhibitors with potent anti-Toxoplasma gondii activity in vitro. J Parasitol 93:694–700. doi: 10.1645/GE-1043R.1. [DOI] [PubMed] [Google Scholar]
  • 78.Jeffers V, Gao H, Checkley LA, Liu Y, Ferdig MT, Sullivan WJ. 2016. Garcinol inhibits GCN5-mediated lysine acetyltransferase activity and prevents replication of the parasite Toxoplasma gondii. Antimicrob Agents Chemother 60:2164–2170. doi: 10.1128/AAC.03059-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Moser MA, Hagelkruys A, Seiser C. 2014. Transcription and beyond: the role of mammalian class I lysine deacetylases. Chromosoma 123:67–78. doi: 10.1007/s00412-013-0441-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sullivan WJ Jr, Jeffers V. 2012. Mechanisms of Toxoplasma gondii persistence and latency. FEMS Microbiol Rev 36:717–733. doi: 10.1111/j.1574-6976.2011.00305.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Shaw MK, He CY, Roos DS, Tilney LG. 2000. Proteasome inhibitors block intracellular growth and replication of Toxoplasma gondii. Parasitology 121:35–47. doi: 10.1017/S0031182099006071. [DOI] [PubMed] [Google Scholar]
  • 82.Skariah S, McIntyre MK, Mordue DG. 2010. Toxoplasma gondii: determinants of tachyzoite to bradyzoite conversion. Parasitol Res 107:253–260. doi: 10.1007/s00436-010-1899-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Yang S, Parmley SF. 1997. Toxoplasma gondii expresses two distinct lactate dehydrogenase homologous genes during its life cycle in intermediate hosts. Gene 184:1–12. doi: 10.1016/S0378-1119(96)00566-5. [DOI] [PubMed] [Google Scholar]
  • 84.Dando C, Schroeder ER, Hunsaker LA, Deck LM, Royer RE, Zhou X, Parmley SF, Vander Jagt DL. 2001. The kinetic properties and sensitivities to inhibitors of lactate dehydrogenases (LDH1 and LDH2) from Toxoplasma gondii: comparisons with pLDH from Plasmodium falciparum. Mol Biochem Parasitol 118:23–32. doi: 10.1016/S0166-6851(01)00360-7. [DOI] [PubMed] [Google Scholar]
  • 85.Chang HR, Pechère JC. 1989. In-vitro toxoplasmacidal activity of cationic electron carriers. J Antimicrob Chemother 23:229–235. doi: 10.1093/jac/23.2.229. [DOI] [PubMed] [Google Scholar]
  • 86.Kadri D, Crater AK, Lee H, Solomon VR, Ananvoranich S. 2014. The potential of quinoline derivatives for the treatment of Toxoplasma gondii infection. Exp Parasitol 145:135–144. doi: 10.1016/j.exppara.2014.08.008. [DOI] [PubMed] [Google Scholar]
  • 87.Strobl JS, Seibert CW, Li Y, Nagarkatti R, Mitchell SM, Rosypal AC, Rathore D, Lindsay DS. 2009. Inhibition of Toxoplasma gondii and Plasmodium falciparum infections in vitro by NSC3852, a redox active antiproliferative and tumor cell differentiation agent. J Parasitol 95:215–223. doi: 10.1645/GE-1608.1. [DOI] [PubMed] [Google Scholar]
  • 88.Portes J de A, Netto CD, da Silva AJM, Costa PRR, DaMatta RA, dos Santos TAT, De Souza W, Seabra SH. 2012. A new type of pterocarpanquinone that affects Toxoplasma gondii tachyzoites in vitro. Vet Parasitol 186:261–269. doi: 10.1016/j.vetpar.2011.11.008. [DOI] [PubMed] [Google Scholar]
  • 89.Fox DT, Schmidt EN, Tian H, Dhungana S, Valentine MC, Warrington NV, Phillips PD, Finney KB, Cope EK, Leid JG, Testa CA, Koppisch AT. 2014. Sub-inhibitory fosmidomycin exposures elicits oxidative stress in Salmonella enterica serovar Typhimurium LT2. PLoS One 9:e95271. doi: 10.1371/journal.pone.0095271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Dunay IR, Chan WC, Haynes RK, Sibley LD. 2009. Artemisone and artemiside control acute and reactivated toxoplasmosis in a murine model. Antimicrob Agents Chemother 53:4450–4456. doi: 10.1128/AAC.00502-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Steyn JD, Wiesner L, du Plessis LH, Grobler AF, Smith PJ, Chan W-C, Haynes RK, Kotzé AF. 2011. Absorption of the novel artemisinin derivatives artemisone and artemiside: potential application of Pheroid™ technology. Int J Pharm 414:260–266. doi: 10.1016/j.ijpharm.2011.05.003. [DOI] [PubMed] [Google Scholar]
  • 92.Nagelschmitz J, Voith B, Wensing G, Roemer A, Fugmann B, Haynes RK, Kotecka BM, Rieckmann KH, Edstein MD. 2008. First assessment in humans of the safety, tolerability, pharmacokinetics, and ex vivo pharmacodynamic antimalarial activity of the new artemisinin derivative artemisone. Antimicrob Agents Chemother 52:3085–3091. doi: 10.1128/AAC.01585-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gaafar MR, Mady RF, Diab RG, Shalaby TI. 2014. Chitosan and silver nanoparticles: promising anti-toxoplasma agents. Exp Parasitol 143:30–38. doi: 10.1016/j.exppara.2014.05.005. [DOI] [PubMed] [Google Scholar]
  • 94.Beverley JK, Fry BA. 1957. Sulphadimidine, pyrimethamine and dapsone in the treatment of toxoplasmosis in mice. Br J Pharmacol Chemother 12:189–193. doi: 10.1111/j.1476-5381.1957.tb00119.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.McFadden DC, Seeber F, Boothroyd JC. 1997. Use of Toxoplasma gondii expressing beta-galactosidase for colorimetric assessment of drug activity in vitro. Antimicrob Agents Chemother 41:1849–1853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Pfefferkorn ER, Pfefferkorn LC. 1977. Specific labeling of intracellular Toxoplasma gondii with uracil. J Protozool 24:449–453. doi: 10.1111/j.1550-7408.1977.tb04774.x. [DOI] [PubMed] [Google Scholar]
  • 97.Gubbels M-J, Li C, Striepen B. 2003. High-throughput growth assay for Toxoplasma gondii using yellow fluorescent protein. Antimicrob Agents Chemother 47:309–316. doi: 10.1128/AAC.47.1.309-316.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Moine E, Denevault-Sabourin C, Debierre-Grockiego F, Silpa L, Gorgette O, Barale J-C, Jacquiet P, Brossier F, Gueiffier A, Dimier-Poisson I, Enguehard-Gueiffier C. 2015. A small-molecule cell-based screen led to the identification of biphenylimidazoazines with highly potent and broad-spectrum anti-apicomplexan activity. Eur J Med Chem 89:386–400. doi: 10.1016/j.ejmech.2014.10.057. [DOI] [PubMed] [Google Scholar]
  • 99.Fomovska A, Huang Q, El Bissati K, Mui EJ, Witola WH, Cheng G, Zhou Y, Sommerville C, Roberts CW, Bettis S, Prigge ST, Afanador GA, Hickman MR, Lee PJ, Leed SE, Auschwitz JM, Pieroni M, Stec J, Muench SP, Rice DW, Kozikowski AP, McLeod R. 2012. Novel N-benzoyl-2-hydroxybenzamide disrupts unique parasite secretory pathway. Antimicrob Agents Chemother 56:2666–2682. doi: 10.1128/AAC.06450-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Stec J, Huang Q, Pieroni M, Kaiser M, Fomovska A, Mui E, Witola WH, Bettis S, McLeod R, Brun R, Kozikowski AP. 2012. Synthesis, biological evaluation, and structure-activity relationships of N-benzoyl-2-hydroxybenzamides as agents active against P. falciparum (K1 strain), trypanosomes, and Leishmania. J Med Chem 55:3088–3100. doi: 10.1021/jm2015183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Doggett JS, Nilsen A, Forquer I, Wegmann KW, Jones-Brando L, Yolken RH, Bordón C, Charman SA, Katneni K, Schultz T, Burrows JN, Hinrichs DJ, Meunier B, Carruthers VB, Riscoe MK. 2012. Endochin-like quinolones are highly efficacious against acute and latent experimental toxoplasmosis. Proc Natl Acad Sci U S A 109:15936–15941. doi: 10.1073/pnas.1208069109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Teo CF, Zhou XW, Bogyo M, Carruthers VB. 2007. Cysteine protease inhibitors block Toxoplasma gondii microneme secretion and cell invasion. Antimicrob Agents Chemother 51:679–688. doi: 10.1128/AAC.01059-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Carey KL, Westwood NJ, Mitchison TJ, Ward GE. 2004. A small-molecule approach to studying invasive mechanisms of Toxoplasma gondii. Proc Natl Acad Sci U S A 101:7433–7438. doi: 10.1073/pnas.0307769101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hall CI, Reese ML, Weerapana E, Child MA, Bowyer PW, Albrow VE, Haraldsen JD, Phillips MR, Sandoval ED, Ward GE, Cravatt BF, Boothroyd JC, Bogyo M. 2011. Chemical genetic screen identifies Toxoplasma DJ-1 as a regulator of parasite secretion, attachment, and invasion. Proc Natl Acad Sci U S A 108:10568–10573. doi: 10.1073/pnas.1105622108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kortagere S. 2012. Screening for small molecule inhibitors of Toxoplasma gondii. Expert Opin Drug Discov 7:1193–1206. doi: 10.1517/17460441.2012.729036. [DOI] [PubMed] [Google Scholar]
  • 106.Chio LC, Queener SF. 1993. Identification of highly potent and selective inhibitors of Toxoplasma gondii dihydrofolate reductase. Antimicrob Agents Chemother 37:1914–1923. doi: 10.1128/AAC.37.9.1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Gangjee A, Jain HD, Phan J, Guo X, Queener SF, Kisliuk RL. 2010. 2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors. Bioorg Med Chem 18:953–961. doi: 10.1016/j.bmc.2009.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bag S, Tawari NR, Queener SF, Degani MS. 2010. Synthesis and biological evaluation of biguanide and dihydrotriazine derivatives as potential inhibitors of dihydrofolate reductase of opportunistic microorganisms. J Enzyme Inhib Med Chem 25:331–339. doi: 10.3109/14756360903179443. [DOI] [PubMed] [Google Scholar]
  • 109.Gangjee A, Adair OO, Pagley M, Queener SF. 2008. N9-substituted 2,4-diaminoquinazolines: synthesis and biological evaluation of lipophilic inhibitors of Pneumocystis carinii and Toxoplasma gondii dihydrofolate reductase. J Med Chem 51:6195–6200. doi: 10.1021/jm800694g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Piper JR, Johnson CA, Krauth CA, Carter RL, Hosmer CA, Queener SF, Borotz SE, Pfefferkorn ER. 1996. Lipophilic antifolates as agents against opportunistic infections. 1. Agents superior to trimetrexate and piritrexim against Toxoplasma gondii and Pneumocystis carinii in in vitro evaluations. J Med Chem 39:1271–1280. [DOI] [PubMed] [Google Scholar]
  • 111.Gangjee A, Jain HD, Queener SF, Kisliuk RL. 2008. The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents. J Med Chem 51:4589–4600. doi: 10.1021/jm800244v. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Allegra CJ, Boarman D, Kovacs JA, Morrison P, Beaver J, Chabner BA, Masur H. 1990. Interaction of sulfonamide and sulfone compounds with Toxoplasma gondii dihydropteroate synthase. J Clin Invest 85:371–379. doi: 10.1172/JCI114448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Gangjee A, Lin X, Biondo LR, Queener SF. 2010. CoMFA analysis of tgDHFR and rlDHFR based on antifolates with 6-5 fused ring system using the all-orientation search (AOS) routine and a modified cross-validated r2-guided region selection (q2-GRS) routine and its initial application. Bioorg Med Chem 18:1684–1701. doi: 10.1016/j.bmc.2009.12.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mui EJ, Jacobus D, Milhous WK, Schiehser G, Hsu H, Roberts CW, Kirisits MJ, McLeod R. 2005. Triazine inhibits Toxoplasma gondii tachyzoites in vitro and in vivo. Antimicrob Agents Chemother 49:3463–3467. doi: 10.1128/AAC.49.8.3463-3467.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Asai T, Takeuchi T, Diffenderfer J, Sibley LD. 2002. Identification of small-molecule inhibitors of nucleoside triphosphate hydrolase in Toxoplasma gondii. Antimicrob Agents Chemother 46:2393–2399. doi: 10.1128/AAC.46.8.2393-2399.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hui R, El Bakkouri M, Sibley LD. 2015. Designing selective inhibitors for calcium-dependent protein kinases in apicomplexans. Trends Pharmacol Sci 36:452–460. doi: 10.1016/j.tips.2015.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Lourido S, Shuman J, Zhang C, Shokat KM, Hui R, Sibley LD. 2010. Calcium-dependent protein kinase 1 is an essential regulator of exocytosis in Toxoplasma. Nature 465:359–362. doi: 10.1038/nature09022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Larson ET, Ojo KK, Murphy RC, Johnson SM, Zhang Z, Kim JE, Leibly DJ, Fox AMW, Reid MC, Dale EJ, Perera BGK, Kim J, Hewitt SN, Hol WGJ, Verlinde CLMJ, Fan E, Van Voorhis WC, Maly DJ, Merritt EA. 2012. Multiple determinants for selective inhibition of apicomplexan calcium-dependent protein kinase CDPK1. J Med Chem 55:2803–2810. doi: 10.1021/jm201725v. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lourido S, Zhang C, Lopez MS, Tang K, Barks J, Wang Q, Wildman SA, Shokat KM, Sibley LD. 2013. Optimizing small molecule inhibitors of calcium-dependent protein kinase 1 to prevent infection by Toxoplasma gondii. J Med Chem 56:3068–3077. doi: 10.1021/jm4001314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Doggett JS, Ojo KK, Fan E, Maly DJ, Van Voorhis WC. 2014. Bumped kinase inhibitor 1294 treats established Toxoplasma gondii infection. Antimicrob Agents Chemother 58:3547–3549. doi: 10.1128/AAC.01823-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Castellanos-Gonzalez A, White AC, Ojo KK, Vidadala RSR, Zhang Z, Reid MC, Fox AMW, Keyloun KR, Rivas K, Irani A, Dann SM, Fan E, Maly DJ, Van Voorhis WC. 2013. A novel calcium-dependent protein kinase inhibitor as a lead compound for treating cryptosporidiosis. J Infect Dis 208:1342–1348. doi: 10.1093/infdis/jit327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ojo KK, Reid MC, Kallur Siddaramaiah L, Müller J, Winzer P, Zhang Z, Keyloun KR, Vidadala RSR, Merritt EA, Hol WGJ, Maly DJ, Fan E, Van Voorhis WC, Hemphill A. 2014. Neospora caninum calcium-dependent protein kinase 1 is an effective drug target for neosporosis therapy. PLoS One 9:e92929. doi: 10.1371/journal.pone.0092929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Ojo KK, Eastman RT, Vidadala R, Zhang Z, Rivas KL, Choi R, Lutz JD, Reid MC, Fox AMW, Hulverson MA, Kennedy M, Isoherranen N, Kim LM, Comess KM, Kempf DJ, Verlinde CLMJ, Su X-Z, Kappe SHI, Maly DJ, Fan E, Van Voorhis WC. 2014. A specific inhibitor of PfCDPK4 blocks malaria transmission: chemical-genetic validation. J Infect Dis 209:275–284. doi: 10.1093/infdis/jit522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kamau E, Meehan T, Lavine MD, Arrizabalaga G, Mustata Wilson G, Boyle J. 2011. A novel benzodioxole-containing inhibitor of Toxoplasma gondii growth alters the parasite cell cycle. Antimicrob Agents Chemother 55:5438–5451. doi: 10.1128/AAC.00455-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Sharma H, Landau MJ, Sullivan TJ, Kumar VP, Dahlgren MK, Jorgensen WL, Anderson KS. 2014. Virtual screening reveals allosteric inhibitors of the Toxoplasma gondii thymidylate synthase-dihydrofolate reductase. Bioorg Med Chem Lett 24:1232–1235. doi: 10.1016/j.bmcl.2013.12.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Boyom FF, Fokou PVT, Tchokouaha LRY, Spangenberg T, Mfopa AN, Kouipou RMT, Mbouna CJ, Donfack VFD, Zollo PHA. 2014. Repurposing the open access malaria box to discover potent inhibitors of Toxoplasma gondii and Entamoeba histolytica. Antimicrob Agents Chemother 58:5848–5854. doi: 10.1128/AAC.02541-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Armande Guiduemde W, Shelat AA, Bouck D, Duffy S, Crowther GJ, Davis PH, Smithson DC, Connelly M, Clark J, Zhu F, Jiménez-Díaz MB, Martinez MS, Wilson EB, Tripathi AK, Gut J, Sharlow ER, Bathurst I, El Mazouni F, Fowble JW, Forquer I, McGinley PL, Castro S, Angulo-Barturen I, Ferrer S, Rosenthal PJ, DeRisi JL, Sullivan DJ, Lazo JS, Roos DS, Riscoe MK, Phillips MA, Rathod PK, Van Voorhis WC, Avery VM, Guy RK. 2010. Chemical genetics of Plasmodium falciparum. Nature 465:311–315. doi: 10.1038/nature09099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Joyce BR, Konrad C, Wek RC, Sullivan WJ Jr. 2011. Translation control is critical during acute and chronic stages of toxoplasmosis infection. Expert Rev Anti Infect Ther 9:1–3. doi: 10.1586/eri.10.146. [DOI] [PubMed] [Google Scholar]
  • 129.Sugi T, Kato K, Kobayashi K, Kurokawa H, Takemae H, Gong H, Recuenco FC, Iwanaga T, Horimoto T, Akashi H. 2011. 1NM-PP1 treatment of mice infected with Toxoplasma gondii. J Vet Med Sci 73:1377–1379. doi: 10.1292/jvms.11-0085. [DOI] [PubMed] [Google Scholar]
  • 130.Vidadala RSR, Rivas KL, Ojo KK, Hulverson MA, Zambriski JA, Bruzual I, Schultz TL, Huang W, Zhang Z, Scheele S, DeRocher AE, Choi R, Barrett LK, Siddaramaiah LK, Hol WGJ, Fan E, Merritt EA, Parsons M, Freiberg G, Marsh K, Kempf DJ, Carruthers VB, Isoherranen N, Doggett JS, Van Voorhis WC, Maly DJ. 2016. Development 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 toxoplasmosis. J Med Chem 59:6531–6546. doi: 10.1021/acs.jmedchem.6b00760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Hupe DJ, Pfefferkorn ER, Behrens ND, Peters K. 1991. L-651,582 inhibition of intracellular parasitic protozoal growth correlates with host-cell directed effects. J Pharmacol Exp Ther 256:462–467. [PubMed] [Google Scholar]
  • 132.Hinshaw JC, Suh D-Y, Garnier P, Buckner FS, Eastman RT, Matsuda SPT, Joubert BM, Coppens I, Joiner KA, Merali S, Nash TE, Prestwich GD. 2003. Oxidosqualene cyclase inhibitors as antimicrobial agents. J Med Chem 46:4240–4243. doi: 10.1021/jm034126t. [DOI] [PubMed] [Google Scholar]
  • 133.Ma C, Tran J, Gu F, Ochoa R, Li C, Sept D, Werbovetz K, Morrissette N. 2010. Dinitroaniline activity in Toxoplasma gondii expressing wild-type or mutant alpha-tubulin. Antimicrob Agents Chemother 54:1453–1460. doi: 10.1128/AAC.01150-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Huang W, Ojo KK, Zhang Z, Rivas K, Vidadala RSR, Scheele S, DeRocher AE, Choi R, Hulverson MA, Barrett LK, Bruzual I, Siddaramaiah LK, Kerchner KM, Kurnick MD, Freiberg GM, Kempf D, Hol WGJ, Merritt EA, Neckermann G, de Hostos EL, Isoherranen N, Maly DJ, Parsons M, Doggett JS, Van Voorhis WC, Fan E. 2015. SAR studies of 5-aminopyrazole-4-carboxamide analogues as potent and selective inhibitors of Toxoplasma gondii CDPK1. ACS Med Chem Lett 6:1184–1189. doi: 10.1021/acsmedchemlett.5b00319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Yadav V, Chu CK, Rais RH, Al Safarjalani ON, Guarcello V, Naguib FNM, el Kouni MH. 2004. Synthesis, biological activity and molecular modeling of 6-benzylthioinosine analogues as subversive substrates of Toxoplasma gondii adenosine kinase. J Med Chem 47:1987–1996. doi: 10.1021/jm030537y. [DOI] [PubMed] [Google Scholar]
  • 136.Rais RH, Al Safarjalani ON, Yadav V, Guarcello V, Kirk M, Chu CK, Naguib FNM, el Kouni MH. 2005. 6-Benzylthioinosine analogues as subversive substrate of Toxoplasma gondii adenosine kinase: activities and selective toxicities. Biochem Pharmacol 69:1409–1419. doi: 10.1016/j.bcp.2005.02.017. [DOI] [PubMed] [Google Scholar]
  • 137.Kim YA, Sharon A, Chu CK, Rais RH, Al Safarjalani ON, Naguib FNM, el Kouni MH. 2007. Synthesis, biological evaluation and molecular modeling studies of N6-benzyladenosine analogues as potential anti-toxoplasma agents. Biochem Pharmacol 73:1558–1572. doi: 10.1016/j.bcp.2007.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Al Safarjalani ON, Rais RH, Kim YA, Chu CK, Naguib FNM, el Kouni MH. 2008. 7-Deaza-6-benzylthioinosine analogues as subversive substrate of Toxoplasma gondii adenosine kinase: activities and selective toxicities. Biochem Pharmacol 76:958–966. doi: 10.1016/j.bcp.2008.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kim YA, Sharon A, Chu CK, Rais RH, Al Safarjalani ON, Naguib FNM, el Kouni MH. 2008. Structure-activity relationships of 7-deaza-6-benzylthioinosine analogues as ligands of Toxoplasma gondii adenosine kinase. J Med Chem 51:3934–3945. doi: 10.1021/jm800201s. [DOI] [PubMed] [Google Scholar]
  • 140.Al Safarjalani ON, Rais RH, Kim YA, Chu CK, Naguib FNM, El Kouni MH. 2010. Carbocyclic 6-benzylthioinosine analogues as subversive substrates of Toxoplasma gondii adenosine kinase: biological activities and selective toxicities. Biochem Pharmacol 80:955–963. doi: 10.1016/j.bcp.2010.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Piper JR, Johnson CA, Hosmer CA, Carter RL, Pfefferkorn ER, Borotz SE, Queener SF. 1993. Lipophilic antifolates as candidates against opportunistic infections. Adv Exp Med Biol 338:429–433. doi: 10.1007/978-1-4615-2960-6_86. [DOI] [PubMed] [Google Scholar]
  • 142.Gangjee A, Vasudevan A, Queener SF, Kisliuk RL. 1996. 2,4-Diamino-5-deaza-6-substituted pyrido[2,3-d]pyrimidine antifolates as potent and selective nonclassical inhibitors of dihydrofolate reductases. J Med Chem 39:1438–1446. doi: 10.1021/jm950786p. [DOI] [PubMed] [Google Scholar]
  • 143.Gangjee A, Adair O, Queener SF. 1999. Pneumocystis carinii and Toxoplasma gondii dihydrofolate reductase inhibitors and antitumor agents: synthesis and biological activities of 2,4-diamino-5-methyl-6-[(monosubstituted anilino)methyl] pyrido[2,3-d]pyrimidines. J Med Chem 42:2447–2455. doi: 10.1021/jm990079m. [DOI] [PubMed] [Google Scholar]
  • 144.Gangjee A, Adair OO, Queener SF. 2003. Synthesis and biological evaluation of 2,4-diamino-6-(arylaminomethyl)pyrido[2,3-d]pyrimidines as inhibitors of Pneumocystis carinii and Toxoplasma gondii dihydrofolate reductase and as antiopportunistic infection and antitumor agents. J Med Chem 46:5074–5082. doi: 10.1021/jm030312n. [DOI] [PubMed] [Google Scholar]
  • 145.Mitchell SM, Zajac AM, Davis WL, Lindsay DS. 2004. Efficacy of ponazuril in vitro and in preventing and treating Toxoplasma gondii infections in mice. J Parasitol 90:639–642. doi: 10.1645/GE-250R. [DOI] [PubMed] [Google Scholar]
  • 146.Kul O, Yildiz K, Ocal N, Freyre A, Deniz A, Karahan S, Atmaca HT, Gokpinar S, Dincel GC, Uzunalioğlu T, Terzi OS. 2013. In-vivo efficacy of toltrazuril on experimentally induced Toxoplasma gondii tissue cysts in lambs: a novel strategy for prevention of human exposure to meat-borne toxoplasmosis. Res Vet Sci 94:269–276. doi: 10.1016/j.rvsc.2012.08.001. [DOI] [PubMed] [Google Scholar]
  • 147.Oz HS. 2014. Maternal and congenital toxoplasmosis, currently available and novel therapies in horizon. Front Microbiol 5:385. doi: 10.3389/fmicb.2014.00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Rosowsky A, Forsch RA, Queener SF. 2002. Inhibition of Pneumocystis carinii, Toxoplasma gondii, and Mycobacterium avium dihydrofolate reductases by 2,4-diamino-5-[2-methoxy-5-(omega-carboxyalkyloxy)benzyl]pyrimidines: marked improvement in potency relative to trimethoprim and species selectivity relative to piritrexim. J Med Chem 45:233–241. doi: 10.1021/jm010407u. [DOI] [PubMed] [Google Scholar]
  • 149.Zeng Y-B, Zhu S-H, Dong H, Han H-Y, Jiang L-L, Wang Q, Cheng J, Zhao Q-P, Ma W-J, Huang B. 2012. Great efficacy of sulfachloropyrazine-sodium against acute murine toxoplasmosis. Asian Pac J Trop Biomed 2:70–75. doi: 10.1016/S2221-1691(11)60193-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Rosowsky A, Papoulis AT, Forsch RA, Queener SF. 1999. Synthesis and antiparasitic and antitumor activity of 2, 4-diamino-6-(arylmethyl)-5,6,7,8-tetrahydroquinazoline analogues of piritrexim. J Med Chem 42:1007–1017. doi: 10.1021/jm980572i. [DOI] [PubMed] [Google Scholar]
  • 151.Gorla SK, Kavitha M, Zhang M, Liu X, Sharling L, Gollapalli DR, Striepen B, Hedstrom L, Cuny GD. 2012. Selective and potent urea inhibitors of Cryptosporidium parvum inosine 5′-monophosphate dehydrogenase. J Med Chem 55:7759–7771. doi: 10.1021/jm3007917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Choi H-J, Yu S-T, Lee K-I, Choi J-K, Chang B-Y, Kim S-Y, Ko M-H, Song H-O, Park H. 2014. 6-Trifluoromethyl-2-thiouracil possesses anti-Toxoplasma gondii effect in vitro and in vivo with low hepatotoxicity. Exp Parasitol 143:24–29. doi: 10.1016/j.exppara.2014.05.002. [DOI] [PubMed] [Google Scholar]
  • 153.Drozdowicz YM, Shaw M, Nishi M, Striepen B, Liwinski HA, Roos DS, Rea PA. 2003. Isolation and characterization of TgVP1, a type I vacuolar H+-translocating pyrophosphatase from Toxoplasma gondii. The dynamics of its subcellular localization and the cellular effects of a diphosphonate inhibitor. J Biol Chem 278:1075–1085. [DOI] [PubMed] [Google Scholar]
  • 154.Ling Y, Sahota G, Odeh S, Chan JMW, Araujo FG, Moreno SNJ, Oldfield E. 2005. Bisphosphonate inhibitors of Toxoplasma gondi growth: in vitro, QSAR, and in vivo investigations. J Med Chem 48:3130–3140. doi: 10.1021/jm040132t. [DOI] [PubMed] [Google Scholar]
  • 155.Shubar HM, Mayer JP, Hopfenmüller W, Liesenfeld O. 2008. A new combined flow-cytometry-based assay reveals excellent activity against Toxoplasma gondii and low toxicity of new bisphosphonates in vitro and in vivo. J Antimicrob Chemother 61:1110–1119. doi: 10.1093/jac/dkn047. [DOI] [PubMed] [Google Scholar]
  • 156.Szajnman SH, García Liñares GE, Li Z-H, Jiang C, Galizzi M, Bontempi EJ, Ferella M, Moreno SNJ, Docampo R, Rodriguez JB. 2008. Synthesis and biological evaluation of 2-alkylaminoethyl-1,1-bisphosphonic acids against Trypanosoma cruzi and Toxoplasma gondii targeting farnesyl diphosphate synthase. Bioorg Med Chem 16:3283–3290. doi: 10.1016/j.bmc.2007.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Rosso VS, Szajnman SH, Malayil L, Galizzi M, Moreno SNJ, Docampo R, Rodriguez JB. 2011. Synthesis and biological evaluation of new 2-alkylaminoethyl-1,1-bisphosphonic acids against Trypanosoma cruzi and Toxoplasma gondii targeting farnesyl diphosphate synthase. Bioorg Med Chem 19:2211–2217. doi: 10.1016/j.bmc.2011.02.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Recher M, Barboza AP, Li Z-H, Galizzi M, Ferrer-Casal M, Szajnman SH, Docampo R, Moreno SNJ, Rodriguez JB. 2013. Design, synthesis and biological evaluation of sulfur-containing 1,1-bisphosphonic acids as antiparasitic agents. Eur J Med Chem 60:431–440. doi: 10.1016/j.ejmech.2012.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Liñares GG, Gismondi S, Codesido NO, Moreno SNJ, Docampo R, Rodriguez JB. 2007. Fluorine-containing aryloxyethyl thiocyanate derivatives are potent inhibitors of Trypanosoma cruzi and Toxoplasma gondii proliferation. Bioorg Med Chem Lett 17:5068–5071. doi: 10.1016/j.bmcl.2007.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Dantas-Leite L, Urbina JA, de Souza W, Vommaro RC. 2005. Antiproliferative synergism of azasterols and antifolates against Toxoplasma gondii. Int J Antimicrob Agents 25:130–135. doi: 10.1016/j.ijantimicag.2004.08.016. [DOI] [PubMed] [Google Scholar]
  • 161.Wei S, Marches F, Daniel B, Sonda S, Heidenreich K, Curiel T. 2002. Pyridinylimidazole p38 mitogen-activated protein kinase inhibitors block intracellular Toxoplasma gondii replication. Int J Parasitol 32:969–977. doi: 10.1016/S0020-7519(02)00061-9. [DOI] [PubMed] [Google Scholar]
  • 162.Maubon D, Bougdour A, Wong Y-S, Brenier-Pinchart M-P, Curt A, Hakimi M-A, Pelloux H. 2010. Activity of the histone deacetylase inhibitor FR235222 on Toxoplasma gondii: inhibition of stage conversion of the parasite cyst form and study of new derivative compounds. Antimicrob Agents Chemother 54:4843–4850. doi: 10.1128/AAC.00462-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.de Lima LPO, Seabra SH, Carneiro H, Barbosa HS. 2015. Effect of 3-bromopyruvate and atovaquone on infection during in vitro interaction of Toxoplasma gondii and LLC-MK2 cells. Antimicrob Agents Chemother 59:5239–5249. doi: 10.1128/AAC.00337-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Khan AA, Nasr M, Araujo FG. 1998. Two 2-hydroxy-3-alkyl-1,4-naphthoquinones with in vitro and in vivo activities against Toxoplasma gondii. Antimicrob Agents Chemother 42:2284–2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Ferreira RA, Oliveira AB, Gualberto SA, Vitor RWA. 2002. Activity of natural and synthetic naphthoquinones against Toxoplasma gondii, in vitro and in murine models of infection. Parasite (Paris) 9:261–269. doi: 10.1051/parasite/2002093261. [DOI] [PubMed] [Google Scholar]
  • 166.Tapia RA, Alegria L, Pessoa CD, Salas C, Cortés MJ, Valderrama JA, Sarciron ME, Pautet F, Walchshofer N, Fillion H. 2003. Synthesis and antiprotozoal activity of naphthofuranquinones and naphthothiophenequinones containing a fused thiazole ring. Bioorg Med Chem 11:2175–2182. doi: 10.1016/S0968-0896(03)00122-6. [DOI] [PubMed] [Google Scholar]
  • 167.Baramee A, Coppin A, Mortuaire M, Pelinski L, Tomavo S, Brocard J. 2006. Synthesis and in vitro activities of ferrocenic aminohydroxynaphthoquinones against Toxoplasma gondii and Plasmodium falciparum. Bioorg Med Chem 14:1294–1302. doi: 10.1016/j.bmc.2005.09.054. [DOI] [PubMed] [Google Scholar]
  • 168.Ferreira RA, Oliveira AB, Ribeiro MFB, Tafuri WL, Vitor RWA. 2006. Toxoplasma gondii: in vitro and in vivo activities of the hydroxynaphthoquinone 2-hydroxy-3-(1′-propen-3-phenyl)-1,4-naphthoquinone alone or combined with sulfadiazine. Exp Parasitol 113:125–129. doi: 10.1016/j.exppara.2005.12.006. [DOI] [PubMed] [Google Scholar]
  • 169.Ferreira RA, de Oliveira AB, Gualberto SA, Miguel Del Corral JM, Fujiwara RT, Gazzinelli Guimarães PH, de Almeida Vitor RW. 2012. New naphthoquinones and an alkaloid with in vitro activity against Toxoplasma gondii RH and EGS strains. Exp Parasitol 132:450–457. doi: 10.1016/j.exppara.2012.09.003. [DOI] [PubMed] [Google Scholar]
  • 170.Smith AT, Livingston MR, Mai A, Filetici P, Queener SF, Sullivan WJ. 2007. Quinoline derivative MC1626, a putative GCN5 histone acetyltransferase (HAT) inhibitor, exhibits HAT-independent activity against Toxoplasma gondii. Antimicrob Agents Chemother 51:1109–1111. doi: 10.1128/AAC.01256-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Paugam A, Creuzet C, Dupouy-Camet J, Roisin P. 2002. In vitro effects of gliotoxin, a natural proteasome inhibitor, on the infectivity and proteolytic activity of Toxoplasma gondii. Parasitol Res 88:785–787. doi: 10.1007/s00436-002-0644-1. [DOI] [PubMed] [Google Scholar]
  • 172.Fox BA, Bzik DJ. 2003. Organisation and sequence determination of glutamine-dependent carbamoyl phosphate synthetase II in Toxoplasma gondii. Int J Parasitol 33:89–96. doi: 10.1016/S0020-7519(02)00214-X. [DOI] [PubMed] [Google Scholar]
  • 173.Tapia RA, Prieto Y, Pautet F, Walchshofer N, Fillion H, Fenet B, Sarciron ME. 2003. Synthesis and antiprotozoal evaluation of benzothiazolopyrroloquinoxalinones, analogues of kuanoniamine A. Bioorg Med Chem 11:3407–3412. doi: 10.1016/S0968-0896(03)00311-0. [DOI] [PubMed] [Google Scholar]
  • 174.Krivogorsky B, Grundt P, Yolken R, Jones-Brando L. 2008. Inhibition of Toxoplasma gondii by indirubin and tryptanthrin analogs. Antimicrob Agents Chemother 52:4466–4469. doi: 10.1128/AAC.00903-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Holmes M, Crater AK, Dhudshia B, Thadani AN, Ananvoranich S. 2011. Toxoplasma gondii: inhibitory activity and encystation effect of securinine and pyrrolidine derivatives on Toxoplasma growth. Exp Parasitol 127:370–375. doi: 10.1016/j.exppara.2010.09.002. [DOI] [PubMed] [Google Scholar]
  • 176.Krivogorsky B, Pernat JA, Douglas KA, Czerniecki NJ, Grundt P. 2012. Structure-activity studies of some berberine analogs as inhibitors of Toxoplasma gondii. Bioorg Med Chem Lett 22:2980–2982. doi: 10.1016/j.bmcl.2012.02.038. [DOI] [PubMed] [Google Scholar]
  • 177.Krivogorsky B, Nelson AC, Douglas KA, Grundt P. 2013. Tryptanthrin derivatives as Toxoplasma gondii inhibitors: structure-activity-relationship of the 6-position. Bioorg Med Chem Lett 23:1032–1035. doi: 10.1016/j.bmcl.2012.12.024. [DOI] [PubMed] [Google Scholar]
  • 178.Leepin A, Stüdli A, Brun R, Stephens CE, Boykin DW, Hemphill A. 2008. Host cells participate in the in vitro effects of novel diamidine analogues against tachyzoites of the intracellular apicomplexan parasites Neospora caninum and Toxoplasma gondii. Antimicrob Agents Chemother 52:1999–2008. doi: 10.1128/AAC.01236-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kropf C, Debache K, Rampa C, Barna F, Schorer M, Stephens CE, Ismail MA, Boykin DW, Hemphill A. 2012. The adaptive potential of a survival artist: characterization of the in vitro interactions of Toxoplasma gondii tachyzoites with di-cationic compounds in human fibroblast cell cultures. Parasitology 139:208–220. doi: 10.1017/S0031182011001776. [DOI] [PubMed] [Google Scholar]
  • 180.Barna F, Debache K, Vock CA, Küster T, Hemphill A. 2013. In vitro effects of novel ruthenium complexes in Neospora caninum and Toxoplasma gondii tachyzoites. Antimicrob Agents Chemother 57:5747–5754. doi: 10.1128/AAC.02446-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Portes JA, Souza TG, dos Santos TA, da Silva LL, Ribeiro TP, Pereira MD, Horn A Jr, Fernandes C, DaMatta RA, de Souza W, Seabra SH. 2015. Reduction of Toxoplasma gondii development due to inhibition of parasite antioxidant enzymes by a dinuclear iron(III) compound. Antimicrob Agents Chemother 59:7374–7386. doi: 10.1128/AAC.00057-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Oliveira CB, Meurer YS, Oliveira MG, Medeiros WM, Silva FO, Brito AC, Pontes Dde L, Andrade-Neto VF. 2014. Comparative study on the antioxidant and anti-Toxoplasma activities of vanillin and its resorcinarene derivative. Molecules 19:5898–5912. doi: 10.3390/molecules19055898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Chang HR, Jefford CW, Pechère JC. 1989. In vitro effects of three new 1,2,4-trioxanes (pentatroxane, thiahexatroxane, and hexatroxanone) on Toxoplasma gondii. Antimicrob Agents Chemother 33:1748–1752. doi: 10.1128/AAC.33.10.1748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.D'Angelo JG, Bordón C, Posner GH, Yolken R, Jones-Brando L. 2009. Artemisinin derivatives inhibit Toxoplasma gondii in vitro at multiple steps in the lytic cycle. J Antimicrob Chemother 63:146–150. doi: 10.1093/jac/dkn451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Jones-Brando L, D'Angelo J, Posner GH, Yolken R. 2006. In vitro inhibition of Toxoplasma gondii by four new derivatives of artemisinin. Antimicrob Agents Chemother 50:4206–4208. doi: 10.1128/AAC.00793-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Schultz TL, Hencken CP, Woodard LE, Posner GH, Yolken RH, Jones-Brando L, Carruthers VB. 2014. A thiazole derivative of artemisinin moderately reduces Toxoplasma gondii cyst burden in infected mice. J Parasitol 100:516–521. doi: 10.1645/13-451.1. [DOI] [PubMed] [Google Scholar]
  • 187.Hencken CP, Jones-Brando L, Bordón C, Stohler R, Mott BT, Yolken R, Posner GH, Woodard LE. 2010. Thiazole, oxadiazole, and carboxamide derivatives of artemisinin are highly selective and potent inhibitors of Toxoplasma gondii. J Med Chem 53:3594–3601. doi: 10.1021/jm901857d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Peuchmaur M, Saïdani N, Botté C, Maréchal E, Vial H, Wong Y-S. 2008. Enhanced antimalarial activity of novel synthetic aculeatin derivatives. J Med Chem 51:4870–4873. doi: 10.1021/jm8007322. [DOI] [PubMed] [Google Scholar]
  • 189.Zuther E, Johnson JJ, Haselkorn R, McLeod R, Gornicki P. 1999. Growth of Toxoplasma gondii is inhibited by aryloxyphenoxypropionate herbicides targeting acetyl-CoA carboxylase. Proc Natl Acad Sci U S A 96:13387–13392. doi: 10.1073/pnas.96.23.13387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Nebois P, Sarciron ME, Bibal B, Bouammali B, Cherkaoui O, Pautet F, Pétavy AF, Walchshofer N, Fillion H. 2000. Quinonic derivatives active against a virulent strain of Toxoplasma gondii. Synthesis of 2-methylfuro[2,3-g]- and [3,2-g]isoquinolinetriones. Bioorg Med Chem Lett 10:871–873. doi: 10.1016/S0960-894X(00)00112-8. [DOI] [PubMed] [Google Scholar]
  • 191.Sarciron M-E, Nebois P, Pautet F, Pétavy A-F, Fillion H, Walchshofer N. 2002. Quinonic derivatives active against Toxoplasma gondii. Parasitol Res 88:969–971. doi: 10.1007/s00436-002-0615-6. [DOI] [PubMed] [Google Scholar]
  • 192.D'Ascenzio M, Bizzarri B, De Monte C, Carradori S, Bolasco A, Secci D, Rivanera D, Faulhaber N, Bordón C, Jones-Brando L. 2014. Design, synthesis and biological characterization of thiazolidin-4-one derivatives as promising inhibitors of Toxoplasma gondii. Eur J Med Chem 86:17–30. doi: 10.1016/j.ejmech.2014.08.046. [DOI] [PubMed] [Google Scholar]
  • 193.Müller J, Limban C, Stadelmann B, Missir AV, Chirita IC, Chifiriuc MC, Nitulescu GM, Hemphill A. 2009. Thioureides of 2-(phenoxymethyl)benzoic acid 4-R substituted: a novel class of anti-parasitic compounds. Parasitol Int 58:128–135. doi: 10.1016/j.parint.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 194.McNulty J, Keskar K, Jenkins HA, Werstiuk NH, Bordón C, Yolken R, Jones-Brando L. 2015. Synthesis of the cyanobacterial metabolite nostodione A, structural studies and potent antiparasitic activity against Toxoplasma gondii. Org Biomol Chem 13:10015–10024. doi: 10.1039/C5OB01506E. [DOI] [PubMed] [Google Scholar]
  • 195.Brown CE, McNulty J, Bordón C, Yolken R, Jones-Brando L. 2016. Enol ethers as carbonyl surrogates in a modification of the Povarov synthesis of 3-aryl quinolines and their anti-Toxoplasma activity. Org Biomol Chem 14:5951–5955. doi: 10.1039/C6OB01083K. [DOI] [PubMed] [Google Scholar]
  • 196.Kamau ET, Srinivasan AR, Brown MJ, Fair MG, Caraher EJ, Boyle JP. 2012. A focused small-molecule screen identifies 14 compounds with distinct effects on Toxoplasma gondii. Antimicrob Agents Chemother 56:5581–5590. doi: 10.1128/AAC.00868-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Winzer P, Müller J, Aguado-Martínez A, Rahman M, Balmer V, Manser V, Ortega-Mora LM, Ojo KK, Fan E, Maly DJ, Van Voorhis WC, Hemphill A. 2015. In vitro and in vivo effects of the bumped kinase inhibitor 1294 in the related cyst-forming apicomplexans Toxoplasma gondii and Neospora caninum. Antimicrob Agents Chemother 59:6361–6374. doi: 10.1128/AAC.01236-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Dittmar AJ, Drozda AA, Blader IJ. 2016. Drug repurposing screening identifies novel compounds that effectively inhibit Toxoplasma gondii growth. mSphere 1(2):e00042-15. doi: 10.1128/mSphere.00042-15. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental material

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES