Next Article in Journal
Influence of the Incubator as Direct Patient Environment on Bacterial Colonization of Neonates
Next Article in Special Issue
An Uninvited Seat at the Dinner Table: How Apicomplexan Parasites Scavenge Nutrients from the Host
Previous Article in Journal
Molecular Variability and Host Distribution of ‘Candidatus Phytoplasma solani’ Strains from Different Geographic Origins
Previous Article in Special Issue
Structural and Functional Insights into the Microtubule Organizing Centers of Toxoplasma gondii and Plasmodium spp.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toxoplasmosis: Current and Emerging Parasite Druggable Targets

1
Department of Biological Sciences, Beirut Arab University, P.O. Box 11-5020, Riad El Solh, Beirut 1107 2809, Lebanon
2
Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon
3
Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon
4
Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2021, 9(12), 2531; https://doi.org/10.3390/microorganisms9122531
Submission received: 27 October 2021 / Revised: 10 November 2021 / Accepted: 10 November 2021 / Published: 7 December 2021
(This article belongs to the Special Issue The Molecular Life of Apicomplexa)

Abstract

:
Toxoplasmosis is a prevalent disease affecting a wide range of hosts including approximately one-third of the human population. It is caused by the sporozoan parasite Toxoplasma gondii (T. gondii), which instigates a range of symptoms, manifesting as acute and chronic forms and varying from ocular to deleterious congenital or neuro-toxoplasmosis. Toxoplasmosis may cause serious health problems in fetuses, newborns, and immunocompromised patients. Recently, associations between toxoplasmosis and various neuropathies and different types of cancer were documented. In the veterinary sector, toxoplasmosis results in recurring abortions, leading to significant economic losses. Treatment of toxoplasmosis remains intricate and encompasses general antiparasitic and antibacterial drugs. The efficacy of these drugs is hindered by intolerance, side effects, and emergence of parasite resistance. Furthermore, all currently used drugs in the clinic target acute toxoplasmosis, with no or little effect on the chronic form. In this review, we will provide a comprehensive overview on the currently used and emergent drugs and their respective parasitic targets to combat toxoplasmosis. We will also abridge the repurposing of certain drugs, their targets, and highlight future druggable targets to enhance the therapeutic efficacy against toxoplasmosis, hence lessening its burden and potentially alleviating the complications of its associated diseases.

1. Introduction

Toxoplasma gondii (T. gondii) is an obligate intracellular protozoan parasite infecting a broad range of animals, including one-third of the world’s human population [1]. Because of its high prevalence in the United States, the Centers for Disease Control and Prevention classified toxoplasmosis among the neglected parasitic infections that require a public health action control [2]. The pathogenesis of T. gondii varies among patients. The acute form of the disease develops a few days after the infection and is asymptomatic in more than 80% of immunocompetent individuals [1,3]. The remaining patients may exhibit general flu-like symptoms, fever, myalgia, and cervical posterior adenopathy, among other symptoms [1]. In certain regions, e.g., French Guiana and Latin America, severe symptoms including fatal pneumonitis, myocarditis, meningoencephalitis, and polymyositis were noted in some immunocompetent patients who contracted atypical strains of T. gondii [4].
Congenital toxoplasmosis occurs in sero-negative pregnant women acquiring T. gondii as primary infection. The parasite crosses the blood–placenta barrier and reaches the fetus [5]. The transmission rate and severity of congenital toxoplasmosis depend on the gestational trimester at which the infection is acquired [6,7]. Transmission rates of 25% are estimated when the infection occurs during the first trimester, while 54 and 65% transmission rates are estimated when infection occurs during the second or third trimesters, respectively [7,8]. Infection of the fetus during the first trimester often leads to abortion. However, in cases of stillbirth, the baby suffers from severe aberrations of the brain (hydrocephalus, intracranial calcifications, deafness, mental retardation, seizures) and the eyes (retinochoroiditis that may lead to blindness (reviewed in [9])). Infection of the fetus during the second or third trimester is less likely to cause abortion; however, retinochoroiditis and learning difficulties may occur after birth [10]. Retinochoroiditis is the most common symptom of ocular toxoplasmosis and predominantly results from an acquired congenital toxoplasmosis. It presents with posterior uveitis, vitritis, focal necrotizing granulomatous retinitis, and reactive granulomatous choroiditis [11]. It is worth noting that the acquisition of toxoplasmosis during pregnancy varies according to regions [12,13], and atypical T. gondii genotypes were identified and led to re-infection of previously sero-positive pregnant women [14], resulting in a more severe congenital disease [15].
Following an acute infection, T. gondii targets the brain and skeletal muscles, where it persists as latent tissue cysts responsible for chronic toxoplasmosis (reviewed in [16]). The switch from the acute to the chronic form is triggered by the host immune response, among other factors (reviewed in [17,18]). Although the direct symptoms of chronic toxoplasmosis are not fully characterized in healthy individuals, chronic toxoplasmosis was regarded as clinically asymptomatic [19]. Yet, the brain immune response triggers inflammation, which disrupts neuronal connectivity and associates with ventricular dilatation [20,21,22]. In addition, chronic toxoplasmosis correlates with several neuropathies [19], behavioral disorders, and cancers [23]. Nonetheless, direct molecular proofs remain to be elucidated [24,25] (Daher et al., in press).
Immunosuppression triggers the reactivation of chronic toxoplasmosis, leading to serious complications and potential fatality [1,26,27,28]. Reactivation was mostly documented in HIV patients or patients treated with immunosuppressive therapies prior to solid organ or hematopoietic stem cell transplantation [18,29,30,31,32,33,34,35]. Indeed, among solid organ transplanted patients, orthotopic heart transplant recipients presented with the highest risk of reactivation of toxoplasmosis, owing to the high propensity of T. gondii cysts in striated muscles [36]. Chemotherapy administration, in particular, rituximab, also triggered the reactivation of toxoplasmosis [37,38,39,40]. HIV patients with reactivated toxoplasmosis manifest predominantly with neurological symptoms including toxoplasmic encephalitis, encephalopathy, meningoencephalitis, headache, seizures, and poor coordination, while transplanted patients exhibit a more disseminated status [4,41].

2. Current Treatment Modalities of Toxoplasmosis

Treatment of toxoplasmosis remains limited to general antiparasitic/antibacterial drugs (reviewed [42,43]). Therapeutic strategies for this parasitic disease vary according to the disease state and the host immune system.
In congenital toxoplasmosis, the combination of pyrimethamine and sulfadiazine is the recommended first-line therapy (Table 1) [44,45,46,47]. Pyrimethamine is an inhibitor of dihydrofolate reductase (DHFR) enzyme, known to block the synthesis of purines and pyrimidines. Sulfadiazine is an inhibitor of dihydropteroate synthase (DHPS). Since T. gondii synthesizes folates de novo [44], this combination exhibits its antiparasitic activity through blocking the biosynthesis of parasitic folate, thus interrupting nucleic acid synthesis and parasite replication. To reduce the harmful side effects, among which is bone marrow myelosuppression, pyrimethamine/sulfadiazine is administered with folinic acid (leucovorin), which is an active metabolite of folic acid and an essential co-enzyme for nucleic acid synthesis [42,48,49]. Yet adverse side effects still present and comprise neutropenia, thrombocytopenia, leukopenia, and teratogenic potential, if administered during the first trimester of pregnancy. In rare cases agranulocytosis, toxic epidermal necrolysis, and hepatic necrosis may also manifest [50,51,52] (Table 1). In pregnant women suspected to have toxoplasmosis, spiramycin is given prophylactically (reviewed in [43,47]) (Table 1). Spiramycin is a macrolide antibiotic that accumulates in and does not cross the placental barrier, hence preventing the materno–fetal transmission of the parasite. Nonetheless, when the diagnosis confirms the infection of the fetus or neonate, spiramycin treatment should be withdrawn and replaced by the conventional treatment pyrimethamine/sulfadiazine/leucovorin [53].
In immunocompetent patients, acute toxoplasmosis is generally asymptomatic; hence, it does not require any therapeutic intervention. Nevertheless, it was reported in immunocompetent patients of certain areas including South America that atypical strains caused severe complications with multivisceral involvement and potentially life-threatening outcome [54,55]. These symptomatic patients, whether presenting with severe symptoms or following ocular involvement or, even if very rare, following a laboratory-acquired infection, are treated with pyrimethamine/sulfadiazine/folinic acid, pyrimethamine/clindamycin/folinic acid, pyrimethamine/folinic acid/atovaquone, pyrimethamine/azithromycin/folinic acid or trimethoprim/sulfamethoxazole (Table 1) (reviewed in [42]). Of note, clindamycin and azithromycin are antibiotics targeting protein synthesis while atovaquone is a quinone antimicrobial medication targeting the mitochondrial electron transport and the mitochondrial cytochrome bc1 complex (Table 1). Attempts to combine pyrimethamine with atovaquone, clindamycin, or azithromycin proved less effective than pyrimethamine/sulfadiazine. The same results were obtained using cotrimoxazole (trimethoprim-sulfamethoxazole) or atovaquone monotherapy [42,56]. The aforementioned treatment modalities spare chronic toxoplasmosis and target only the acute form of the infection [48,57,58,59]. Reactivation of tissue cysts, which are the hallmark of chronic toxoplasmosis, occurs when the host immunity is suppressed [18,29,30,31,32,33,34]. For instance, in HIV patients an association between CD4 counts and T. gondii-related neurologic symptoms was reported [27]. Reactivation becomes a concern if CD4 counts fall below 200 cells/µL and is the consequence of decreased IFN-γ and cytokine production. In these patients, toxoplasmic encephalitis is the major clinical manifestation and leads to fatality if left untreated [60]. Symptomatic HIV patients revealing fever and dizziness as part of their Toxoplasma encephalitis prodrome pursued medical care quicker than those who did not present with these symptoms, leading to rapid therapeutic intervention, hence reduced rates of mortality [61]. Induction treatment with pyrimethamine/sulfadiazine resulted in 80% response rates [62]. However, severe adverse effects were encountered ranging from fever to skin rash to hematologic complications (leukopenia and thrombocytopenia). Thus, leucovorin was used to overcome these side effects (Table 1) [62]. Atovaquone monotherapy was not effective; nevertheless, induction therapy using the combination of sulfadiazine/pyrimethamine/atovaquone led to high response rates (reviewed in [63]). Anti-retroviral therapy (ART) increased the survival and decreased the mortality and relapse rates in HIV patients with opportunistic infections including reactivation of toxoplasmosis [64,65,66,67,68,69]. In transplant patients, disseminated toxoplasmosis occurs frequently [4,41]; therefore, prophylaxis or even empirical initiation of treatment is recommended in suspected cases and before confirming diagnosis. In a retrospective study, it was demonstrated that the combination of sulfadiazine/pyrimethamine marginally improved survival in hematopoietic stem cell transplantation [70]. In solid organ transplant recipients, where reactivation of chronic toxoplasmosis occurs either as a manifestation derived from an infected donor or occasionally as a primary acquired infection following transplantation, a standard and effective treatment is still lacking, and the therapeutic modalities that are adopted are similar to those used in HIV patients (Table 1) (reviewed in [42,43]).

3. Drug Resistance in Toxoplasma gondii Infections

A gold standard treatment for toxoplasmosis is still lacking. In addition to the multiple reported side effects of clinically used drugs, the emergence of resistance strains was described [48,58,71,72]. This resistance may be one of the factors dictating the failure of treatment in patients with acute toxoplasmosis or in patients who relapse in the course of suppressive therapy. However, the extent at which this drug resistance leads to the failure of treatment cannot be determined easily because of the complex recovery of T. gondii from infected patients [48]. Nonetheless, alterations in the common enzyme targets of the folate pathway, dhfr and dhps, were described [73,74,75,76]. In that sense, in a sulfadiazine-resistant Brazilian isolate from newborns with congenital toxoplasmosis, a large number of polymorphisms were identified in the dhps gene. Yet, no association was found between the profile of susceptibility to sulfadiazine and any identified dhps variants [77]. Three clinically isolated strains (the atypical RMS-2001-MAU strain, the type I B1 strain, and the Type II RMS-1995-ABE strain) were also shown to be resistant to sulfadiazine [78]. Using a proteomic approach, 31 proteins were differentially modulated between sulfadiazine-sensitive and resistant strains of T. gondii, of which the rhoptry protein ROP2A virulence factor was highly abundant in two naturally resistant Type II strains, TgH32045 and TgH32006 [79]. To overcome these problems, continuous efforts to develop promising drug candidates are being made [59,80].

4. Emerging Therapeutic Targets in Toxoplasma gondii Infections

Despite all prophylactic approaches to prevent the infection with T. gondii, an available human vaccine is still out of reach. While the search for a vaccine has been highly pursued, currently used drugs target only the acute form of the disease. Chronic toxoplasmosis, which represents the more prevalent form and associates with dreadful clinical outcomes, reaching fatality in immunocompromised patients, remains an unmet medical need. An ideal drug against toxoplasmosis should affect multiple stages of the parasite life cycle (i.e., tachyzoites responsible for acute toxoplasmosis and bradyzoites responsible for chronic toxoplasmosis). Furthermore, these drugs should (1) target the parasite biology, (2) exhibit low toxicity and tolerable side effects, (3) have high bioavailability, and (4) cross the blood–brain barrier and reach the brain, where the propensity for neuronal cysts is high [81]. A number of preclinical studies were conducted in vitro and prolonged mice survival in vivo (reviewed in [82]). A comprehensive summary of tested drugs and compounds over a decade extending from 2006 to 2016 reported 80 clinically available drugs and a large number of new compounds with more than 40 mechanisms of action. Several target-based drug screens were also identified. These include different kinases, mitochondrial electron transport chain, fatty acid synthesis, DNA synthesis, and replication, among several others [59]. In the following sections, we will provide a comprehensive overview of the different parasite targets and their corresponding emerging drugs (Figure 1, Table 2).

4.1. Targeting the Apicoplast

In apicomplexan parasites, the apicoplast is a nonphotosynthetic organelle formed following endosymbiosis of a green algae. This organelle assumes several functions including the biosynthesis of fatty acids, lipoic acid, and isoprenoids, among other metabolites [83]. The absence of apicoplasts in mammalian cells made them an excellent therapeutic target, and several attempts to target their parasitic functions have been tested (Figure 1, Table 2). These therapeutic targets span several apicoplast enzymes involved in fatty acid synthesis and metabolism. These include the fatty acid synthase II (FASII) [84], the acetyl CoA carboxylase (ACC) that catalyzes the formation of Malonyl-CoA, and the β-ketoacyl ACP synthase III (FabH) [85,86,87]. Clodinafop, thiolactomycin, and triclosan proved efficient in targeting these enzymes and blocking the parasite fatty acid synthesis [85,86].
Another apicoplast therapeutic drug target is the isoprenoid synthesis pathway. Isoprenoid is a precursor of ubiquinone and sterols, playing an important role in cell signaling. 1-deoxy-D-xylulose-5-phosphate (DOXP) was identified in the apicoplast and plays a role in the biosynthesis of isoprenoid [88]. Two key enzymes featured in the DOXP pathway, the DXP reducto-isomerase and DXP synthase, were identified in apicomplexan parasites including T. gondii and are absent in humans, posturing these enzymes as attractive therapeutic drug targets [88]. Fosmidomycin antibiotic proved efficient against these two enzymes; however, resistance problems, high concentrations, and low bioavailability hindered its activity [89,90].
Because of its prokaryotic nature, the apicoplast’s DNA is circular, and its unwinding and supercoiling during replication is controlled and mediated by DNA topo-isomerases including the DNA gyrase [91,92,93]. This enzyme is absent in humans and was the target of several quinolone and fuoroquinolone antibiotics. These include ciprofloxacin, trovafloxacin, ofloxacin and temafloxacin, which exhibited in vitro and in vivo efficacy against T. gondii infections [94,95]. In addition to DNA replication, the prokaryotic nature of the apicoplast also confers a mechanism of protein synthesis independent of that occurring in the nucleus of the parasite. Indeed, the apicoplast encodes proteins and RNA indispensable to its ribosomes, hence its specific proteins [96]. Clindamycin, spiramycin, and azithromycin, known to affect prokaryotic protein synthesis, exhibited toxoplasmicidal activities in vitro and in vivo [97].

4.2. Targeting the Invasion Complex

4.2.1. Microneme Organelles

Micronemes are small rod-shaped organelles of the apical complex of T. gondii. They play a chief role in attachment, gliding, motility, and egress during the invasion process required for parasite survival [98,99,100]. Owing to their uniqueness in apicomplexan parasites, their absence in mammalian cells, and their pivotal role in invasion, micronemes and especially their calcium-mediated secretion have been at the core of parasite targeting. In that sense, T. gondii calcium-dependent protein kinase 1 (TgCDPK1), a parasite cytosolic serine/threonine-protein kinase regulating the calcium-dependent pathway, is essential for micronemal protein secretion. The inhibition of this enzyme impairs host cell invasion capacity [101,102,103]. Several bumped kinase inhibitors (BKIs) selectively inhibited TgCDPK1 [104,105] (Figure 1, Table 2). BKI-1294, a pyrazolo-pyrimidine-based compound, resulted in high inhibition of invasion in vitro and high efficiency against acute toxoplasmosis in vivo when given orally [105]. This same inhibitor also proved efficient against congenital toxoplasmosis in a murine model [106]. The promising efficacy of BKI-1294 was hindered by its cardiac toxicity, halting its clinical development [107,108,109]. Consequently, BKI-1294 was chemically modified by maintaining its TgCDPK1 selectivity and efficacy and reducing its cardiac toxicity. Compound 32 was thus developed and proved efficacious in vitro and in vivo, in particular, through reducing brain cysts [108]. Other pyrazolo-pyrimidine inhibitors of TgCDPK1 were also tested. Another compound (called compound 24) exhibited in vitro nanomolar and submicromolar activity on TgCDPK1 and inhibited parasite proliferation [110]. This BKI analog showed an excellent oral bioavailability, reduced acute and chronic toxoplasmosis in mice, and, more importantly, delayed reactivation of the chronic disease following immunosuppression [110]. Recently, BKI-1748, a 5-aminopyrazole-4-carboxamide compound inhibited proliferation of Neospora caninum and Toxoplasma gondii in vitro. The safety of this compound was tested in zebrafish with no embryonic impairment up to 10 μM, and in pregnant mice, with no pregnancy interference at a dose of 20 mg/kg/day. The efficacy of BKI-1748 was assessed in standardized pregnant mouse models infected with tachyzoites or oocysts of T. gondii and resulted in increased pup survival and profound inhibition of vertical transmission [111]. SP230, an imidazo[1,2-b]pyridazine salt targeting TgCDPK1, proved efficient against murine acute toxoplasmosis in mice. Importantly, administration of SP230 yielded significant efficacy against congenital toxoplasmosis. SP230 resulted in the reduction of parasite burden in 97% of fetuses [112].

4.2.2. Rhoptry Organelles

Rhoptries are organelles of the apical complex discharging their content during parasite invasion. Their protein content plays several roles including contributing to the formation of moving junction and the parasitophorous vacuole membrane (PVM). Furthermore, other functions were unraveled, and some rhoptry proteins play roles as virulence factors, others manipulate host signaling, while others play a role in immune evasion [113,114,115].
ROP2 family contains a group of proteins, with some members sharing more than 70% identity, while other members are structurally more divergent [116]. While the members of this family evolved with all the elements to be active kinases, some members (ROP2, ROP4, ROP7, ROP5) lost some key motifs or residues in the kinase activity domain over time to acquire other functions [116,117]. For instance, ROP2 contributes [118], but is not the only factor [119], to the recruitment of the host mitochondria around the PVM. ROP5 exhibits an inverted topology in the PVM as compared to other members of the family [120], and protein forms a complex with ROP17 and ROP18 (which retained their kinase activity), hence controlling the virulence in mice [121,122]. In that sense, ROP5 and ROP18 allele combinations are tightly related to T. gondii virulence [122,123,124,125], and ROP5 teams up with ROP18 and complements its activity to inhibit the accumulation of the IFN-γ-induced immunity-related GTPases (IRGs) in vivo, hence contributing to the pathogenesis and immune evasion [126]. Owing to the role of ROP5 and ROP18 in virulence, attempts to use this complex as a vaccine strategy were promising in mice [127]. In addition, recombinant ROP5 and ROP18 were evaluated for their diagnostic potential in human toxoplasmosis [128]. ROP16 and ROP18 were also proven as virulence factors through targeting the host cell nucleus and exhibiting their kinase activity to phosphorylate key proteins involved in cell cycle and different signaling pathways [129]. ROP18 is expressed in genotypes I/II demonstrating their role in controlling the virulence of the parasite [130], and transfection of the virulent ROP18 allele into a nonpathogenic type III strain confers virulence and enhances mortality in vivo [131]. Through its kinase activity, ROP18 phosphorylates GTPases, promoting macrophage survival and virulence [132] and ensuring an immune evasion strategy for virulent strains [133]. ROP16, on the other hand, is expressed in genotypes I/III and also plays a key role in the virulence of the parasite [130]. ROP16 phosphorylates STAT3 and STAT6 [134], hence downregulating IL-12, which plays a chief role in mounting an immune response against T. gondii infection [130]. ROP16 also suppresses T cell activity, hence ensuring immune cell evasion [135]. Moreover, direct phosphorylation of STAT3 by ROP16 mimics the IL-10 activity and downregulates IFN-γ, hence enhancing the virulence of T. gondii [134]. Recently, ROP16-mediated activation of STAT6 proved important for type III T. gondii survival through suppression of host cell reactive oxygen species production [136]. Moreover, ROP16 kinase activity silences the cyclin B1 gene promoter, hijacking the function of the host cell epigenetic machinery [137]. The role of ROP proteins in the virulence of the parasite makes them excellent drug target candidates to combat toxoplasmosis. A high-throughput screen to identify small molecule inhibitors of ROP18 identified several inhibitors belonging to oxindoles, 6-azaquinazolines, and pyrazolopyridines chemical scaffolds. Treatment of IFN-γ-activated cells with one of these inhibitors enhanced immunity-related GTPase recruitment to wild type parasites [138]. Thiazolidinone derivatives inhibited T. gondii in vitro, and in silico analysis demonstrated that the best binding affinity of these derivatives was observed in the active site of kinase proteins with a possible effect of one derivative in the active site of ROP18 [139] (Figure 1, Table 2).

4.3. Targeting the Parasite Mitochondrial Electron Transport Pathway

In apicomplexan parasites, the mitochondrial electron transport chain is of central importance for energy production [140]. This complex, present in the mitochondrial electron transport chain, was targeted by several mitochondrial inhibitors, hindering cell respiration and leading to parasite death (Figure 1, Table 2). Atovaquone, clinically used in the treatment and prophylaxis of toxoplasmosis, is an inhibitor of the hydroquinone oxidation site of the bc1 complex [48]. Emerging resistance of the parasite limited its use [141]. Different quinolone derivatives including the endochin-like quinolones (ELQs), which target the hydroquinone reduction site of bc1, have been developed. ELQ-271 and ELQ-316 inhibited parasite growth at nanomolar concentrations in vitro and reduced the number of brain cysts in murine models [142,143,144]. Another compound, ELQ-400, alleviated the burden of acute toxoplasmosis in mice and demonstrated 100% cure rates upon infection of mice with a type I lethal strain [143,145].
Naphthoquinones bind to the hydroquinone oxidation site of the bc1 complex. Seven naphthoquinones exhibited an anti-T. gondii inhibitory effect in vitro. Three out of seven (para-hydroxynaphthoquinones) were able to enhance survival of mice following infection with a virulent T. gondii strain (Ferreira et al., 2002) (Figure 1, Table 2).

4.4. Targeting the Interconversion between Tachyzoites and Bradyzoites

Histone acetylase (HAT) and histone deacetylase (HDAC) enzymes controlling histone acetylation regulate and control the parasite gene expression during the back and forth interconversion between acute and chronic toxoplasmosis. Targeting these enzymes is a plausible therapeutic scenario. The cyclopeptide FR235222, a TgHDAC3 inhibitor, induced in vitro conversion to bradyzoites and inhibited parasite growth [146]. To decrease the toxicity of FR235222, W363 and W399 derivatives were generated and exhibited equivalent IC50 to the mother compound in vitro [147].
Rolipram, a phosphodiestrase-4 (PDE4) inhibitor interfered with the interconversion from tachyzoites to bradyzoites through immunomodulatory activities and significantly reduced the cyst burden in the brains of chronically infected mice [148]. Guanabenz, an FDA-approved antihypertensive drug, interferes with translational control in tachyzoite and bradyzoite stages through inhibition of dephosphorylation of T. gondii eukaryotic initiation factor 2 (TgeIF2). This inhibitor protected mice against acute toxoplasmosis and reduced the brain cyst numbers in chronically infected mice [81].
T. gondii mitogen-activated protein kinase (MAPK) regulates parasite proliferation, response to stress, and stage differentiation. Pyridinylimidazole inhibited TgMAPK1, caused morphological changes, and reduced the virulence of T. gondii [149,150,151]. In conclusion, targeting the interconversion between tachyzoites and bradyzoites can be a promising therapeutic approach.
Heat shock proteins (HSPs) promote host cell invasion, parasite growth, survival, as well as stage conversion from tachyzoite to bradyzoite, hence from the acute to the chronic form of infection [152,153]. HSP60 and 70 are important in the development and survival of T. gondii. While HSP60 is responsible for stage-specific induction of the respiratory pathway, HSP70 plays a role in stage differentiation and virulence [154]. HSP70 protects the parasite from the host immune system. Treatment of mice with quercetin and oligonucleotide reduced HSP70 expression in a virulent T. gondii strain [155]. The 3D structures for T. gondii Hsp60 and Hsp70 were performed by homology modeling, and a virtual screening of 1560 compounds from the NCI Diversity Set III was analyzed and demonstrated that the major exhibited interactions were hydrogen bonding and hydrophobic interactions in binding to HSP60 and HSP70, providing guidelines for the development of inhibitors for these parasitic heat shock proteins [156].

5. Drug Repositioning: A Promising Approach against T. Gondii

Drug repositioning became an advantageous approach to explore more clinical usages of existing drugs. This strategy offers multiple advantages, including the established pharmacokinetic and pharmacodynamic properties of drugs, their assumed targets, reduced cost and timeline, hence accelerating their clinical applications.
Since T. gondii and Plasmodia spp. belong to the phylum Apicomplexa, several anti-malarial drugs were tested against T. gondii. The piperazine acetamide MMV007791, a potent anti-malarial drug, demonstrated the highest efficacy and selectivity against T. gondii [157]. This drug, along with six out of 400 blood stage-active anti-Plasmodium compounds, was screened from the open access Medicines for Malaria Venture Malaria Box and exhibited toxoplasmicidal efficacy [157]. A screen of a wider open access library of 400 compounds, referred to as the Pathogen box, in which several drugs tested pre-clinically against other apicomplexan parasites including Crysptosporidium parvum and Neopsora caninum, revealed the efficacy of 18 compounds against T. gondii [158]. Among these, the anti-Neospora caninum buparvaquone and the anti-Crysptosporidium parvum MMV675968 targeting the mitochondrial electron transport and DHFR, respectively, exhibited a similar targeted activity in T. gondii [158]. In another screen of a chemical compound library by the Drug Discovery Initiative at Japan, two inhibitors, tanshinone IIA (with potential anti-cancer activity) and hydroxyzine (a first-generation antihistamine drug), were identified and seemed to target intermediately differentiated bradyzoites [159].
Tetraoxane, an anti-cancer molecule, significantly prolonged survival of acutely infected mice as compared to control mice, highlighting a potential use of this molecule against toxoplasmosis [160]. Miltefosine, extensively used currently in the treatment of visceral leishmaniasis among other protozoal infections, was not efficient against acute toxoplasmosis but reduced brain cyst burden in chronically infected mice [161]. More recently, the ability of a collection of 666 FDA-approved compounds (Selleck New Compound Library) to inhibit Toxoplasma growth was screened. A total of 68 compounds proved effective and inhibited parasite growth, out of which two compounds, NVP-AEW541 and GSK-J4 HCl, inhibited tachyzoite invasion and proliferation by halting cell cycle progression from G1 to S phase, respectively. Both compounds prolonged survival of acutely infected mice with T. gondii and remarkedly reduced the parasite burden of tissues [162]. Another screen encompassed the ability of 1120 compounds to reduce Toxoplasma replication. A total of 94 compounds, including inhibitors of dopamine or estrogen signaling, blocked parasite replication. Tamoxifen, an established inhibitor of the estrogen receptor, also reduced parasite invasion and replication [163].
Furthermore, a broad spectrum of known DNA damage inducers were evaluated as anti-toxoplasmic drug targets (reviewed in [164]). These genotoxic drugs include topoisomerase-2 inhibitors such as daunorubicin [165], trovafloxacin [95], enrofloxacin [166], and gatifloxacin [167], which proved effective against T. gondii. Topoisomerase-1 inhibitors, including artemisinin, a highly potent anti-malarial dug, and several of its derivatives, mostly artemisone and artemiside, reduced brain cyst burden in chronically infected mice [48,168] and prolonged survival of mice following reactivation of toxoplasmosis [169]. Artemether, with similar topoisomerase-1 inhibitory activities, also exhibited toxoplasmicidal activities [168,170,171]. Moreover, other topoisomerase-1 inhibitors including harmane, harmine, and nor-harmane Top1 inhibitors proved efficient against T. gondii [48]. DNA-intercalating agents including fluphenazine, thioridazine, trifluoperazine, hycantone [159,172], phleomycin [173], mitomycin C [165], the ribonucleotide reductase inhibitors thiosemicarbazones and hydroxyurea [174], the oxidative DNA damage/DNA binding resveratrol [165], and valproic acid [175] also exhibited an anti-Toxoplasma activity (Figure 1, Table 2).
The incessant advances and the enhanced understanding of the immunopathogenesis of toxoplasmosis gave new insights into counteracting immune damages. Hence, immunomodulation strategies have proved efficient to combat acute and, more importantly, chronic toxoplasmosis. In that sense, imiquimod, an FDA-approved immune-modulatory drug for topical use against some viral infections [176] and against cutaneous leishmaniasis [176,177,178,179,180], was explored in murine models of acute and chronic toxoplasmosis. During acute toxoplasmosis, imiquimod treatment reduced the brain cyst burden and impaired the infectivity of the remaining ones. More importantly, treatment of chronically infected mice with imiquimod significantly abridged the number of brain cysts and led to delayed or abortion of reactivation upon immunosuppression [181]. At the molecular level, imiquimod upregulated the expression of toll-like receptors and activated the MyD88 pathway, resulting in the induction of the immune response to control reactivation [181]. Ursolic acid, a natural pentacyclic triterpenoid compound [182], inhibited T. gondii survival, downregulated several proteins of the apical complex including ROP18 and MIC8, and augmented the production of NO, ROS, IL-10, IL-12, GM-CSF, and IFN-γ, while reducing the expression of IL-1, IL-6, TNF-α, TGF-βin T. gondii-infected immune cells, highlighting the immune-modulatory activities of this natural compound [183,184].

6. Concluding Remarks

Despite its prevalence, toxoplasmosis remains a neglected disease of human and veterinary importance. Clinically available drugs target acute but not chronic toxoplasmosis. These drugs are associated with a spectrum of adverse side effects. Moreover, emergence of resistance in parasites was documented. Identifying new drug targets is paramount in creating avenues for new treatment modalities to overcome the drawbacks of these clinically used drugs and improve their pharmacokinetics, bioavailability in target organs (eyes, placenta, fetal compartment, etc.), and, more importantly, their access to the brain after crossing the blood–brain barrier. It is worth noting that chronic toxoplasmosis resides in the brain and skeletal muscles and is associated with severe complications in immunocompromised patients. Ideally, drug candidates should be cost-effective and should act on acute replicating tachyzoite and latent bradyzoite stages, hence preventing acute disease (including newly acquired infection, ocular and congenital toxoplasmosis) and allowing the resolution of chronic toxoplasmosis, its associated diseases, and, more prominently, its reactivation. Several drug targets including unique organelles of the parasite (apicoplast, apical complex organelles (rhoptries and micronemes)) were identified. Drug repurposing was also applied and deemed a useful approach to combat toxoplasmosis. Under this category fall several anti-malarial drugs, anti-cancer and genotoxic drugs, and, more importantly, immunomodulatory drugs that activate the immune response, triggering a toxoplasmicidal outcome. Recent advances in high-throughput sequencing, modeling, and proteomics techniques should help in identifying parasite markers that do not target host factors, hence increasing their specificity and lessening their toxicity. There is also a surge in developing quick diagnostic tests to augment the clinicians’ awareness of reactivation in immunocompromised patients. Finally, because of the importance of the host immune system in controlling the persistence and the back and forth switch between acute and chronic toxoplasmosis, targeting the parasite and enhancing the host immune system against it should be considered as simultaneous approaches to eradicate different forms of the infection. This urgency to develop effective drugs against toxoplasmosis has gained a new dimension following the plethora of research avenues that have associated this infection with major primary neuropathies, behavioral and psychiatric disorders, and some cancers.

Author Contributions

R.E.H., L.T., S.I., M.H., J.E. and M.E.S. contributed to the writing of different sections in this review. H.E.H. thoroughly corrected the review, added appropriate sections, summarized data in Table 1 and Table 2 and in Figure 1. All authors have read and agreed to the published version of the manuscript.

Funding

The principal investigator would like to thank the core support from the Medical Practice Plan (Faculty of Medicine, American University of Beirut) and the American University of Beirut and the Centre National de Recherche Scientifique Libanais (AUB-CNRS-L GRP) funds allocated to her research on Toxoplasma gondii.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Montoya, J.G.; Liesenfeld, O. Toxoplasmosis. Lancet 2004, 363, 1965–1976. [Google Scholar] [CrossRef]
  2. Ben-Harari, R.R.; Connolly, M.P. High burden and low awareness of toxoplasmosis in the United States. Postgrad. Med. 2019, 131, 103–108. [Google Scholar] [CrossRef] [PubMed]
  3. Reza Yazdani, M.; Mehrabi, Z.; Ataei, B.; Baradaran Ghahfarokhi, A.; Moslemi, R.; Pourahmad, M. Frequency of sero-positivity in household members of the patients with positive toxoplasma serology. Rev. Esp. Quimioter. Publ. Of. Soc. Esp. Quimioter. 2018, 31, 506–510. [Google Scholar]
  4. Robert-Gangneux, F.; Dardé, M.-L. Epidemiology of and Diagnostic Strategies for Toxoplasmosis. Clin. Microbiol. Rev. 2012, 25, 264. [Google Scholar] [CrossRef] [Green Version]
  5. Lindsay, D.S.; Dubey, J.P. Toxoplasma gondii: The changing paradigm of congenital toxoplasmosis. Parasitology 2011, 138, 1829–1831. [Google Scholar] [CrossRef] [Green Version]
  6. Yamamoto, L.; Targa, L.S.; Sumita, L.M.; Shimokawa, P.T.; Rodrigues, J.C.; Kanunfre, K.A.; Okay, T.S. Association of Parasite Load Levels in Amniotic Fluid With Clinical Outcome in Congenital Toxoplasmosis. Obstet. Gynecol. 2017, 130, 335–345. [Google Scholar] [CrossRef]
  7. Robbins, J.R.; Zeldovich, V.B.; Poukchanski, A.; Boothroyd, J.C.; Bakardjiev, A.I. Tissue barriers of the human placenta to infection with Toxoplasma gondii. Infect. Immun. 2012, 80, 418–428. [Google Scholar] [CrossRef] [Green Version]
  8. McAuley, J.B. Congenital Toxoplasmosis. J. Pediatr. Infect. Dis. Soc. 2014, 3 (Suppl. 1), S30–S35. [Google Scholar] [CrossRef]
  9. Singh, S. Congenital toxoplasmosis: Clinical features, outcomes, treatment, and prevention. Trop. Parasitol. 2016, 6, 113–122. [Google Scholar] [CrossRef] [Green Version]
  10. Weiss, L.M.; Dubey, J.P. Toxoplasmosis: A history of clinical observations. Int. J. Parasitol. 2009, 39, 895–901. [Google Scholar] [CrossRef] [Green Version]
  11. Vasconcelos-Santos, D.V.; Dodds, E.M.; Orefice, F. Review for disease of the year: Differential diagnosis of ocular toxoplasmosis. Ocul. Immunol. Inflamm. 2011, 19, 171–179. [Google Scholar] [CrossRef]
  12. Nahouli, H.; El Arnaout, N.; Chalhoub, E.; Anastadiadis, E.; El Hajj, H. Seroprevalence of Anti-Toxoplasma gondii Antibodies Among Lebanese Pregnant Women. Vector Borne Zoonotic Dis. 2017, 17, 785–790. [Google Scholar] [CrossRef]
  13. Nowakowska, D.; Colón, I.; Remington, J.S.; Grigg, M.; Golab, E.; Wilczynski, J.; Sibley, L.D. Genotyping of Toxoplasma gondii by Multiplex PCR and Peptide-Based Serological Testing of Samples from Infants in Poland Diagnosed with Congenital Toxoplasmosis. J. Clin. Microbiol. 2006, 44, 1382. [Google Scholar] [CrossRef] [Green Version]
  14. Galal, L.; Sarr, A.; Cuny, T.; Brouat, C.; Coulibaly, F.; Sembène, M.; Diagne, M.; Diallo, M.; Sow, A.; Hamidović, A.; et al. The introduction of new hosts with human trade shapes the extant distribution of Toxoplasma gondii lineages. PLoS Negl. Trop. Dis. 2019, 13, e0007435. [Google Scholar] [CrossRef] [Green Version]
  15. Delhaes, L.; Ajzenberg, D.; Sicot, B.; Bourgeot, P.; Darde, M.L.; Dei-Cas, E.; Houfflin-Debarge, V. Severe congenital toxoplasmosis due to a Toxoplasma gondii strain with an atypical genotype: Case report and review. Prenat. Diagn. 2010, 30, 902–905. [Google Scholar] [CrossRef]
  16. Schlüter, D.; Barragan, A. Advances and Challenges in Understanding Cerebral Toxoplasmosis. Front. Immunol. 2019, 10, 242. [Google Scholar] [CrossRef]
  17. Blanchard, N.; Dunay, I.R.; Schlüter, D. Persistence of Toxoplasma gondii in the central nervous system: A fine-tuned balance between the parasite, the brain and the immune system. Parasite Immunol. 2015, 37, 150–158. [Google Scholar] [CrossRef]
  18. Matta, S.K.; Rinkenberger, N.; Dunay, I.R.; Sibley, L.D. Toxoplasma gondii infection and its implications within the central nervous system. Nat. Rev. Microbiol. 2021, 19, 467–480. [Google Scholar] [CrossRef]
  19. Madireddy, S.; Rivas Chacon, E.D.; Mangat, R. Toxoplasmosis; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2021. [Google Scholar]
  20. Evans, A.K.; Strassmann, P.S.; Lee, I.P.; Sapolsky, R.M. Patterns of Toxoplasma gondii cyst distribution in the forebrain associate with individual variation in predator odor avoidance and anxiety-related behavior in male Long–Evans rats. Brain Behav. Immun. 2014, 37, 122–133. [Google Scholar] [CrossRef] [Green Version]
  21. Hermes, G.; Ajioka, J.W.; Kelly, K.A.; Mui, E.; Roberts, F.; Kasza, K.; Mayr, T.; Kirisits, M.J.; Wollmann, R.; Ferguson, D.J.; et al. Neurological and behavioral abnormalities, ventricular dilatation, altered cellular functions, inflammation, and neuronal injury in brains of mice due to common, persistent, parasitic infection. J. Neuroinflamm. 2008, 5, 48. [Google Scholar] [CrossRef] [Green Version]
  22. Xiao, J.; Li, Y.; Gressitt, K.L.; He, H.; Kannan, G.; Schultz, T.L.; Svezhova, N.; Carruthers, V.B.; Pletnikov, M.V.; Yolken, R.H.; et al. Severance Cerebral complement C1q activation in chronic Toxoplasma infection. Brain Behav. Immun. 2016, 58, 52–56. [Google Scholar] [CrossRef] [Green Version]
  23. Ngô, H.M.; Zhou, Y.; Lorenzi, H.; Wang, K.; Kim, T.K.; Zhou, Y.; El Bissati, K.; Mui, E.; Fraczek, L.; Rajagopala, S.V.; et al. Toxoplasma Modulates Signature Pathways of Human Epilepsy, Neurodegeneration & Cancer. Sci. Rep. 2017, 7, 11496. [Google Scholar]
  24. Johnson, H.J.; Koshy, A.A. Latent Toxoplasmosis Effects on Rodents and Humans: How Much is Real and How Much is Media Hype? mBio 2020, 11, e02164-19. [Google Scholar] [CrossRef] [Green Version]
  25. Johnson, S.K.; Johnson, P.T.J. Toxoplasmosis: Recent Advances in Understanding the Link Between Infection and Host Behavior. Annu. Rev. Anim. Biosci. 2021, 9, 249–264. [Google Scholar] [CrossRef]
  26. Bannoura, S.; El Hajj, R.; Khalifeh, I.; El Hajj, H. Acute disseminated encephalomyelitis and reactivation of cerebral toxoplasmosis in a child: Case report. IDCases 2018, 13, e00434. [Google Scholar] [CrossRef]
  27. Basavaraju, A. Toxoplasmosis in HIV infection: An overview. Trop. Parasitol. 2016, 6, 129–135. [Google Scholar] [CrossRef] [Green Version]
  28. Kodym, P.; MalÝ, M.; Beran, O.; Jilich, D.; Rozsypal, H.; Machala, L.; Holub, M. Incidence, immunological and clinical characteristics of reactivation of latent Toxoplasma gondii infection in HIV-infected patients. Epidemiol. Infect. 2015, 143, 600–607. [Google Scholar] [CrossRef] [Green Version]
  29. Gay, J.; Gendron, N.; Verney, C.; Joste, V.; Dardé, M.L.; Loheac, C.; Vrtovsnik, F.; Argy, N.; Houze, S. Disseminated toxoplasmosis associated with hemophagocytic syndrome after kidney transplantation: A case report and review. Transpl. Infect. Dis. 2019, 21, e13154. [Google Scholar] [CrossRef]
  30. Kollu, V.; Magalhaes-Silverman, M.; Tricot, G.; Ince, D. Toxoplasma Encephalitis following Tandem Autologous Hematopoietic Stem Cell Transplantation: A Case Report and Review of the Literature. Case Rep. Infect. Dis. 2018, 2018, 9409121. [Google Scholar] [CrossRef]
  31. Paccoud, O.; Guitard, J.; Labopin, M.; Surgers, L.; Malard, F.; Battipaglia, G.; Duléry, R.; Hennequin, C.; Mohty, M.; Brissot, E. Features of Toxoplasma gondii reactivation after allogeneic hematopoietic stem-cell transplantation in a high seroprevalence setting. Bone Marrow Transplant. 2020, 55, 93–99. [Google Scholar] [CrossRef]
  32. Ramanan, P.; Scherger, S.; Benamu, E.; Bajrovic, V.; Jackson, W.; Hage, C.A.; Hakki, M.; Baddley, J.W.; Abidi, M.Z. Toxoplasmosis in non-cardiac solid organ transplant recipients: A case series and review of literature. Transpl. Infect. Dis. 2020, 22, e13218. [Google Scholar] [CrossRef] [PubMed]
  33. Ramchandar, N.; Pong, A.; Anderson, E. Identification of disseminated toxoplasmosis by plasma next-generation sequencing in a teenager with rapidly progressive multiorgan failure following haploidentical stem cell transplantation. Pediatr. Blood Cancer 2020, 67, e28205. [Google Scholar] [CrossRef] [PubMed]
  34. Robert-Gangneux, F.; Meroni, V.; Dupont, D.; Botterel, F.; Garcia, J.M.A.; Brenier-Pinchart, M.-P.; Accoceberry, I.; Akan, H.; Abbate, I.; Boggian, K.; et al. Toxoplasmosis in Transplant Recipients, Europe, 2010–2014. Emerg. Infect. Dis. 2018, 24, 1497–1504. [Google Scholar] [CrossRef] [PubMed]
  35. Adekunle, R.O.; Sherman, A.; Spicer, J.O.; Messina, J.A.; Steinbrink, J.M.; Sexton, M.E.; Lyon, G.M.; Mehta, A.K.; Phadke, V.K.; Woodworth, M.H. Clinical characteristics and outcomes of toxoplasmosis among transplant recipients at two US academic medical centers. Transpl. Infect. Dis. 2021, 23, e13636. [Google Scholar] [CrossRef] [PubMed]
  36. La Hoz, R.M.; Morris, M.I.; Infectious Diseases Community of Practice of the American Society of Transplantation. Infectious Diseases Community of Practice of the American Society of Tissue and blood protozoa including toxoplasmosis, Chagas disease, leishmaniasis, Babesia, Acanthamoeba, Balamuthia, and Naegleria in solid organ transplant recipients- Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin. Transplant. 2019, 33, e13546. [Google Scholar] [PubMed]
  37. Holland, M.S.; Sharma, K.; Lee, B.C. Cerebral toxoplasmosis after rituximab therapy for splenic marginal zone lymphoma: A case report and review of the literature. JMM Case Rep. 2015, 2, e005010. [Google Scholar] [CrossRef] [Green Version]
  38. Lee, E.B.; Ayoubi, N.; Albayram, M.; Kariyawasam, V.; Motaparthi, K. Cerebral toxoplasmosis after rituximab for pemphigus vulgaris. JAAD Case Rep. 2019, 6, 37–41. [Google Scholar] [CrossRef] [Green Version]
  39. Morjaria, S.; Epstein, D.J.; Romero, F.A.; Taur, Y.; Seo, S.K.; Papanicolaou, G.A.; Hatzoglou, V.; Rosenblum, M.; Perales, M.-A.; Scordo, M.; et al. Toxoplasma Encephalitis in Atypical Hosts at an Academic Cancer Center. Open Forum Infect. Dis. 2016, 3, ofw070. [Google Scholar] [CrossRef] [Green Version]
  40. Safa, G.; Darrieux, L. Cerebral Toxoplasmosis After Rituximab Therapy. JAMA Intern. Med. 2013, 173, 924–926. [Google Scholar] [CrossRef] [Green Version]
  41. Rajapakse, S.; Weeratunga, P.; Rodrigo, C.; de Silva, N.L.; Fernando, S.D. Prophylaxis of human toxoplasmosis: A systematic review. Pathog. Glob. Health 2017, 111, 333–342. [Google Scholar] [CrossRef]
  42. Dunay, I.R.; Gajurel, K.; Dhakal, R.; Liesenfeld, O.; Montoya, J.G. Treatment of Toxoplasmosis: Historical Perspective, Animal Models, and Current Clinical Practice. Clin. Microbiol. Rev. 2018, 31, e00057-17. [Google Scholar] [CrossRef] [Green Version]
  43. Konstantinovic, N.; Guegan, H.; Stajner, T.; Belaz, S.; Robert-Gangneux, F. Treatment of toxoplasmosis: Current options and future perspectives. Food Waterborne Parasitol. 2019, 15, e00036. [Google Scholar] [CrossRef]
  44. Blume, M.; Seeber, F. Metabolic interactions between Toxoplasma gondii and its host. F1000Research 2018, 7, 1719. [Google Scholar] [CrossRef] [Green Version]
  45. Lapinskas, P.J.; Ben-Harari, R.R. Perspective on current and emerging drugs in the treatment of acute and chronic toxoplasmosis. Postgrad. Med. 2019, 131, 589–596. [Google Scholar] [CrossRef]
  46. Remington, J.S.; Thulliez, P.; Montoya, J.G. Recent Developments for Diagnosis of Toxoplasmosis. J. Clin. Microbiol. 2004, 42, 941. [Google Scholar] [CrossRef] [Green Version]
  47. Silva, D.R.; Sardi, J.D.C.O.; Freires, I.A.; Silva, A.C.B.; Rosalen, P.L. In silico approaches for screening molecular targets in Candida albicans: A proteomic insight into drug discovery and development. Eur. J. Pharmacol. 2019, 842, 64–69. [Google Scholar] [CrossRef]
  48. Alday, P.H.; Doggett, J.S. Drugs in development for toxoplasmosis: Advances, challenges, and current status. Drug Des. Dev. Ther. 2017, 11, 273–293. [Google Scholar] [CrossRef] [Green Version]
  49. Katlama, C.; Mouthon, B.; Gourdon, D.; Lapierre, D.; Rousseau, F. Atovaquone as long-term suppressive therapy for toxoplasmic encephalitis in patients with AIDS and multiple drug intolerance. Atovaquone Expanded Access Group. Aids 1996, 10, 1107–1112. [Google Scholar]
  50. Ben-Harari, R.R.; Goodwin, E.; Casoy, J. Adverse Event Profile of Pyrimethamine-Based Therapy in Toxoplasmosis: A Systematic Review. Drugs R D 2017, 17, 523–544. [Google Scholar] [CrossRef] [Green Version]
  51. Ardabili, S.; Kohl, J.; Gul, G.; Hodel, M. What obstetricians should be aware of: Serious side effects of antibiotic toxoplasmosis treatment in pregnancy. BMJ Case Rep. 2021, 14, e240809. [Google Scholar] [CrossRef]
  52. Shammaa, A.M.; Powell, T.G.; Benmerzouga, I. Adverse outcomes associated with the treatment of Toxoplasma infections. Sci. Rep. 2021, 11, 1035. [Google Scholar] [CrossRef] [PubMed]
  53. Paquet, C.; Yudin, M.H. No. 285-Toxoplasmosis in Pregnancy: Prevention, Screening, and Treatment. J. Obstet. Gynaecol. Can. 2018, 40, e687–e693. [Google Scholar] [CrossRef] [PubMed]
  54. Demar, M.P.; Ajzenberg, D.; Maubon, D.; Djossou, F.; Panchoe, D.; Punwasi, W.; Valery, N.; Peneau, C.; Daigre, J.; Aznar, C.; et al. Fatal outbreak of human toxoplasmosis along the Maroni River: Epidemiological, clinical, and parasitological aspects. Clin. Infect. Dis. 2007, 45, e88–e95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Demar, M.; Hommel, D.; Djossou, F.; Peneau, C.; Boukhari, R.; Louvel, D.; Bourbigot, A.M.; Nasser, V.; Ajzenberg, D.; Darde, M.L.; et al. Acute toxoplasmoses in immunocompetent patients hospitalized in an intensive care unit in French Guiana. Clin. Microbiol. Infect. 2012, 18, E221–E331. [Google Scholar] [CrossRef] [Green Version]
  56. Rajapakse, S.; Chrishan Shivanthan, M.; Samaranayake, N.; Rodrigo, C.; Deepika Fernando, S. Antibiotics for human toxoplasmosis: A systematic review of randomized trials. Pathog. Glob. Health 2013, 107, 162–169. [Google Scholar] [CrossRef] [Green Version]
  57. Schmidt, D.R.; Hogh, B.; Andersen, O.; Hansen, S.H.; Dalhoff, K.; Petersen, E. Treatment of infants with congenital toxoplasmosis: Tolerability and plasma concentrations of Sulfadiazine and Pyrimethamine. Eur. J. Pediatr. 2006, 165, 19–25. [Google Scholar] [CrossRef]
  58. Montazeri, M.; Mehrzadi, S.; Sharif, M.; Sarvi, S.; Tanzifi, A.; Aghayan, S.A.; Daryani, A. Drug Resistance in Toxoplasma gondii. Front. Microbiol. 2018, 9, 2587. [Google Scholar] [CrossRef] [Green Version]
  59. Montazeri, M.; Sharif, M.; Sarvi, S.; Mehrzadi, S.; Ahmadpour, E.; Daryani, A. A Systematic Review of In vitro and In vivo Activities of Anti-Toxoplasma Drugs and Compounds (2006–2016). Front. Microbiol. 2017, 8, 25. [Google Scholar] [CrossRef] [Green Version]
  60. Luft, B.J.; Remington, J.S. Toxoplasmic Encephalitis in AIDS. Clin. Infect. Dis. 1992, 15, 211–222. [Google Scholar] [CrossRef]
  61. Li, Y.; Zeng, Y.-M.; Liu, M.; Lu, Y.-Q.; Liu, X.-Y.; Zhang, Y.-L.; Jiang, Z.-S.; Yang, T.-T.; Sun, Y.; Lan, K.; et al. Development of a risk scoring system for prognostication in HIV-related toxoplasma encephalitis. BMC Infect. Dis. 2020, 20, 923. [Google Scholar] [CrossRef]
  62. Porter, S.B.; Sande, M.A. Toxoplasmosis of the central nervous system in the acquired immunodeficiency syndrome. N. Engl. J. Med. 1992, 327, 1643–1648. [Google Scholar] [CrossRef]
  63. Wei, H.X.; Wei, S.S.; Lindsay, D.S.; Peng, H.J. A Systematic Review and Meta-Analysis of the Efficacy of Anti-Toxoplasma gondii Medicines in Humans. PLoS ONE 2015, 10, e0138204. [Google Scholar]
  64. Podzamczer, D.; Miró, J.M.; Ferrer, E.; Gatell, J.M.; Ramón, J.M.; Ribera, E.; Sirera, G.; Cruceta, A.; Knobel, H.; Domingo, P.; et al. Thrice-weekly Sulfadiazine-Pyrimethamine for maintenance therapy of toxoplasmic encephalitis in HIV-infected patients. Spanish Toxoplasmosis Study Group. Eur. J. Clin. Microbiol. Infect. Dis. 2000, 19, 89–95. [Google Scholar] [CrossRef]
  65. Vidal, J.E.; Hernandez, A.V.; de Oliveira, A.C.; Dauar, R.F.; Barbosa, S.P., Jr.; Focaccia, R. Cerebral toxoplasmosis in HIV-positive patients in Brazil: Clinical features and predictors of treatment response in the HAART era. AIDS Patient Care STDS 2005, 19, 626–634. [Google Scholar] [CrossRef]
  66. Connolly, M.P.; Goodwin, E.; Schey, C.; Zummo, J. Toxoplasmic encephalitis relapse rates with Pyrimethamine-based therapy: Systematic review and meta-analysis. Pathog. Glob. Health 2017, 111, 31–44. [Google Scholar] [CrossRef]
  67. Connolly, M.P.; Haitsma, G.; Hernández, A.V.; Vidal, J.E. Systematic review and meta-analysis of secondary prophylaxis for prevention of HIV-related toxoplasmic encephalitis relapse using trimethoprim-sulfamethoxazole. Pathog. Glob. Health 2017, 111, 327–331. [Google Scholar] [CrossRef] [Green Version]
  68. Mayor, A.M.; Santos, D.M.F.; Dworkin, M.S.; Ríos-Olivares, E.; Hunter-Mellado, R.F. Toxoplasmic encephalitis in an AIDS cohort at Puerto Rico before and after highly active antiretroviral therapy (HAART). Am. J. Trop. Med. Hyg. 2011, 84, 838–841. [Google Scholar] [CrossRef]
  69. Martin-Iguacel, R.; Ahlström, M.G.; Touma, M.; Engsig, F.N.; Stærke, N.B.; Stærkind, M.; Obel, N.; Rasmussen, L.D. Incidence, presentation and outcome of toxoplasmosis in HIV infected in the combination antiretroviral therapy era. J. Infect. 2017, 75, 263–273. [Google Scholar] [CrossRef]
  70. Mele, A.; Paterson, P.J.; Prentice, H.G.; Leoni, P.; Kibbler, C.C. Toxoplasmosis in bone marrow transplantation: A report of two cases and systematic review of the literature. Bone Marrow Transplant. 2002, 29, 691–698. [Google Scholar] [CrossRef] [Green Version]
  71. Robert-Gangneux, F.; Binisti, P.; Antonetti, D.; Brezin, A.; Yera, H.; Dupouy-Camet, J. Usefulness of immunoblotting and Goldmann-Witmer coefficient for biological diagnosis of toxoplasmic retinochoroiditis. Eur. J. Clin. Microbiol. Infect. Dis. 2004, 23, 34–38. [Google Scholar] [CrossRef]
  72. Rodríguez-Díaz, J.C.; Martínez-Grueiro, M.M.; Martínez-Fernández, A.R. Comparative activity of several antibiotics against Toxoplasma gondii in a mouse model. Enferm. Infect. Microbiol. Clin. 1993, 11, 543–546. [Google Scholar]
  73. Doliwa, C.; Escotte-Binet, S.; Aubert, D.; Sauvage, V.; Velard, F.; Schmid, A.; Villena, I. Sulfadiazine resistance in Toxoplasma gondii: No involvement of overexpression or polymorphisms in genes of therapeutic targets and ABC transporters. Parasite 2013, 20, 19. [Google Scholar] [CrossRef] [Green Version]
  74. Doliwa, C.; Escotte-Binet, S.; Aubert, D.; Velard, F.; Schmid, A.; Geers, R.; Villena, I. Induction of Sulfadiazine resistance in vitro in Toxoplasma gondii. Exp. Parasitol. 2013, 133, 131–136. [Google Scholar] [CrossRef]
  75. Oliveira, C.; Meurer, Y.S.; Andrade, J.; Costa, M.E.; Andrade, M.; Silva, L.A.; Lanza, D.C.; Vítor, R.W.; Andrade-Neto, V.F. Pathogenicity and phenotypic Sulfadiazine resistance of Toxoplasma gondii isolates obtained from livestock in northeastern Brazil. Mem. Inst. Oswaldo Cruz 2016, 111, 391–398. [Google Scholar] [CrossRef] [Green Version]
  76. Aspinall, T.V.; Joynson, D.H.; Guy, E.; Hyde, J.E.; Sims, P.F. The molecular basis of sulfonamide resistance in Toxoplasma gondii and implications for the clinical management of toxoplasmosis. J. Infect. Dis. 2002, 185, 1637–1643. [Google Scholar] [CrossRef] [Green Version]
  77. Silva, L.A.; Reis-Cunha, J.L.; Bartholomeu, D.C.; Vitor, R.W. Genetic Polymorphisms and Phenotypic Profiles of Sulfadiazine-Resistant and Sensitive Toxoplasma gondii Isolates Obtained from Newborns with Congenital Toxoplasmosis in Minas Gerais, Brazil. PLoS ONE 2017, 12, e0170689. [Google Scholar] [CrossRef]
  78. Meneceur, P.; Bouldouyre, M.A.; Aubert, D.; Villena, I.; Menotti, J.; Sauvage, V.J.; Garin, F.; Derouin, F. In vitro susceptibility of various genotypic strains of Toxoplasma gondii to Pyrimethamine, Sulfadiazine, and Atovaquone. Antimicrob. Agents Chemother. 2008, 52, 1269–1277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Doliwa, C.; Xia, D.; Escotte-Binet, S.; Newsham, E.L.; Aubert, D.; Randle, N.; Wastling, J.M.; Villena, I. Identification of differentially expressed proteins in Sulfadiazine resistant and sensitive strains of Toxoplasma gondii using difference-gel electrophoresis (DIGE). Int. J. Parasitol. Drugs Drug Resist. 2013, 3, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Escotte-Binet, S.; Huguenin, A.; Aubert, D.; Martin, A.P.; Kaltenbach, M.; Florent, I.; Villena, I. Metallopeptidases of Toxoplasma gondii: In silico identification and gene expression. Parasite 2018, 25, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Benmerzouga, I.; Checkley, L.A.; Ferdig, M.T.; Arrizabalaga, G.; Wek, R.C.; Sullivan, W.J., Jr. Guanabenz repurposed as an antiparasitic with activity against acute and latent toxoplasmosis. Antimicrob. Agents Chemother. 2015, 59, 6939–6945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. McFarland, M.M.; Zach, S.J.; Wang, X.; Potluri, L.-P.; Neville, A.; Vennerstrom, J.L.; Davis, P.H. Review of Experimental Compounds Demonstrating Anti-Toxoplasma Activity. Antimicrob. Agents Chemother. 2016, 60, 7017–7034. [Google Scholar] [CrossRef] [Green Version]
  83. Saremy, S.; Boroujeni, M.E.; Bhattacharjee, B.; Mittal, V.; Chatterjee, J. Identification of potential apicoplast associated therapeutic targets in human and animal pathogen Toxoplasma gondii ME49. Bioinformation 2011, 7, 379–383. [Google Scholar] [CrossRef] [Green Version]
  84. Sonda, S.; Hehl, A.B. Lipid biology of Apicomplexa: Perspectives for new drug targets, particularly for Toxoplasma gondii. Trends Parasitol. 2006, 22, 41–47. [Google Scholar] [CrossRef]
  85. Waller, R.; Keeling, P.; Donald, R.G.K.; Striepen, B.; Handman, E.; Lang-Unnasch, N.; Cowman, A.F.; Besra, G.; Roos, D.; McFadden, G.I. Nuclear-encoded proteins target to the plastid in Toxoplasma gondii and Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 1998, 95, 12352–12357. [Google Scholar] [CrossRef] [Green Version]
  86. Zuther, E.; Johnson, J.J.; Haselkorn, R.; McLeod, R.; Gornicki, P. Growth of Toxoplasma gondii is inhibited by aryloxyphenoxypropionate herbicides targeting acetyl-CoA carboxylase. Proc. Natl. Acad. Sci. USA 1999, 96, 13387–13392. [Google Scholar] [CrossRef] [Green Version]
  87. Seeber, F.; Soldati-Favre, D. Metabolic pathways in the apicoplast of apicomplexa. Int. Rev. Cell Mol. Biol. 2010, 281, 161–228. [Google Scholar]
  88. Jomaa, H.; Wiesner, J.; Sanderbrand, S.; Altincicek, B.; Weidemeyer, C.; Hintz, M.; Turbachova, I.; Eberl, M.; Zeidler, J.; Lichtenthaler, H.K.; et al. Inhibitors of the nonmevalonate pathway of isoprenoid biosynthesis as antimalarial drugs. Science 1999, 285, 1573–1576. [Google Scholar] [CrossRef] [Green Version]
  89. Ling, Y.; Sahota, G.; Odeh, S.; Chan, J.M.; Araujo, F.G.; Moreno, S.N.; Oldfield, E. Bisphosphonate inhibitors of Toxoplasma gondi growth: In vitro, QSAR, and in vivo investigations. J. Med. Chem. 2005, 48, 3130–3140. [Google Scholar] [CrossRef]
  90. Clastre, M.; Goubard, A.; Prel, A.; Mincheva, Z.; Viaud-Massuart, M.-C.; Bout, D.; Rideau, M.; Velge-Roussel, F.; Laurent, F. The methylerythritol phosphate pathway for isoprenoid biosynthesis in coccidia: Presence and sensitivity to fosmidomycin. Exp. Parasitol. 2007, 116, 375–384. [Google Scholar] [CrossRef]
  91. Garcia-Estrada, C.; Prada, C.F.; Fernandez-Rubio, C.; Rojo-Vazquez, F.; Balana-Fouce, R. DNA topoisomerases in apicomplexan parasites: Promising targets for drug discovery. Proc. Biol. Sci. 2010, 277, 1777–1787. [Google Scholar] [CrossRef] [Green Version]
  92. Maxwell, A. DNA gyrase as a drug target. Biochem. Soc. Trans. 1999, 27, 48–53. [Google Scholar] [CrossRef] [Green Version]
  93. McFadden, G.I.; Roos, D.S. Apicomplexan plastids as drug targets. Trends Microbiol. 1999, 7, 328–333. [Google Scholar] [CrossRef]
  94. Gozalbes, R.; Brun-Pascaud, M.; Garcia-Domenech, R.; Galvez, J.; Girard, P.M.; Doucet, J.P.; Derouin, F. Anti-toxoplasma activities of 24 quinolones and fluoroquinolones in vitro: Prediction of activity by molecular topology and virtual computational techniques. Antimicrob. Agents Chemother. 2000, 44, 2771–2776. [Google Scholar] [CrossRef] [Green Version]
  95. Khan, A.A.; Slifer, T.; Araujo, F.G.; Remington, J.S. Trovafloxacin is active against Toxoplasma gondii. Antimicrob. Agents Chemother. 1996, 40, 1855–1859. [Google Scholar] [CrossRef] [Green Version]
  96. Reiff, S.B.; Vaishnava, S.; Striepen, B. The HU protein is important for apicoplast genome maintenance and inheritance in Toxoplasma gondii. Eukaryot. Cell 2012, 11, 905–915. [Google Scholar] [CrossRef] [Green Version]
  97. Pfefferkorn, E.R.; Nothnagel, R.F.; Borotz, S.E. Parasiticidal effect of Clindamycin on Toxoplasma gondii grown in cultured cells and selection of a drug-resistant mutant. Antimicrob. Agents Chemother. 1992, 36, 1091–1096. [Google Scholar] [CrossRef] [Green Version]
  98. Dubremetz, J.; Garcia-Réguet, N.; Conseil, V.; Fourmaux, M.N. Apical organelles and host-cell invasion by Apicomplexa. Int. J. Parasitol. 1998, 28, 1007–1013. [Google Scholar] [CrossRef]
  99. Langsley, G.; Heussler, V.; Chaussepied, M.; Stanway, R.R.; Lüder, C.G.K. Intracellular survival of apicomplexan parasites and host cell modification. Int. J. Parasitol. 2009, 39, 163–173. [Google Scholar]
  100. Portes, J.; Barrias, E.; Travassos, R.; Attias, M.; De Souza, W. Toxoplasma gondii Mechanisms of Entry Into Host Cells. Front. Cell. Infect. Microbiol. 2020, 10, 294. [Google Scholar] [CrossRef]
  101. Cardew, E.M.; Verlinde, C.L.M.J.; Pohl, E. The calcium-dependent protein kinase 1 from Toxoplasma gondii as target for structure-based drug design. Parasitology 2018, 145, 210–218. [Google Scholar] [CrossRef] [Green Version]
  102. Murphy, R.C.; Ojo, K.K.; Larson, E.T.; Castellanos-Gonzalez, A.; Perera, B.G.; Keyloun, K.R.; Kim, J.E.; Bhandari, J.G.; Muller, N.R.; Verlinde, C.L.; et al. Discovery of Potent and Selective Inhibitors of Calcium-Dependent Protein Kinase 1 (CDPK1) from C. parvum and T. gondii. ACS Med. Chem. Lett. 2010, 1, 331–335. [Google Scholar] [CrossRef] [PubMed]
  103. Ojo, K.K.; Larson, E.T.; Keyloun, K.R.; Castaneda, L.J.; DeRocher, A.E.; Inampudi, K.K.; E Kim, J.; Arakaki, T.L.; Murphy, R.C.; Zhang, L.; et al. Toxoplasma gondii calcium-dependent protein kinase 1 is a target for selective kinase inhibitors. Nat. Struct. Mol. Biol. 2010, 17, 602–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Winzer, P.; Müller, J.; Aguado-Martínez, A.; Rahman, M.; Balmer, V.; Manser, V.; Ortega-Mora, L.M.; Ojo, K.K.; Fan, E.; Maly, D.J.; et al. 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. 2015, 59, 6361–6374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Doggett, J.S.; Ojo, K.K.; Fan, E.; Maly, D.; Van Voorhis, W.C. Bumped kinase inhibitor 1294 treats established Toxoplasma gondii infection. Antimicrob. Agents Chemother. 2014, 58, 3547–3549. [Google Scholar] [CrossRef] [Green Version]
  106. Müller, J.; Aguado-Martínez, A.; Ortega-Mora, L.M.; Moreno-Gonzalo, J.; Ferre, I.; Hulverson, M.A.; Choi, R.; McCloskey, M.C.; Barrett, L.K.; Maly, D.J.; et al. Development of a murine vertical transmission model for Toxoplasma gondii oocyst infection and studies on the efficacy of bumped kinase inhibitor (BKI)-1294 and the naphthoquinone buparvaquone against congenital toxoplasmosis. J. Antimicrob. Chemother. 2017, 72, 2334–2341. [Google Scholar] [CrossRef] [Green Version]
  107. Schaefer, D.A.; Betzer, D.P.; Smith, K.D.; Millman, Z.G.; Michalski, H.C.; Menchaca, S.E.; Zambriski, J.A.; Ojo, K.K.; Hulverson, M.A.; Arnold, S.L.M.; et al. Novel Bumped Kinase Inhibitors Are Safe. and Effective Therapeutics in the Calf Clinical Model. for Cryptosporidiosis. J. Infect. Dis. 2016, 214, 1856–1864. [Google Scholar] [CrossRef]
  108. Vidadala, R.S.R.; Rivas, K.L.; Ojo, K.K.; Hulverson, M.A.; Zambriski, J.A.; Bruzual, I.; Schultz, T.L.; Huang, W.; Zhang, Z.; Scheele, S.; et al. 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. 2016, 59, 6531–6546. [Google Scholar] [CrossRef] [Green Version]
  109. Vandenberg, J.I.; Perry, M.D.; Perrin, M.J.; Mann, S.A.; Ke, Y.; Hill, A.P. hERG K(+) channels: Structure, function, and clinical significance. Physiol. Rev. 2012, 92, 1393–1478. [Google Scholar] [CrossRef] [Green Version]
  110. Rutaganira, F.U.; Barks, J.; Dhason, M.S.; Wang, Q.; Lopez, M.S.; Long, S.; Radke, J.B.; Jones, N.G.; Maddirala, A.R.; Janetka, J.W.; et al. Inhibition of Calcium Dependent Protein Kinase 1 (CDPK1) by Pyrazolopyrimidine Analogs Decreases Establishment and Reoccurrence of Central Nervous System Disease by Toxoplasma gondii. J. Med. Chem. 2017, 60, 9976–9989. [Google Scholar] [CrossRef]
  111. Imhof, D.; Anghel, N.; Winzer, P.; Balmer, V.; Ramseier, J.; Hänggeli, K.; Choi, R.; Hulverson, M.A.; Whitman, G.R.; Arnold, S.L.; et al. In vitro activity, safety and in vivo efficacy of the novel bumped kinase inhibitor BKI-1748 in non-pregnant and pregnant mice experimentally infected with Neospora caninum tachyzoites and Toxoplasma gondii oocysts. Int. J. Parasitol. Drugs Drug Resist. 2021, 16, 90–101. [Google Scholar] [CrossRef]
  112. Débare, H.; Moiré, N.; Baron, F.; Lantier, L.; Héraut, B.; Van Langendonck, N.; Denevault-Sabourin, C.; Dimier-Poisson, I.; Debierre-Grockiego, F. A Novel Calcium-Dependent Protein Kinase 1 Inhibitor Potently Prevents Toxoplasma gondii Transmission to Foetuses in Mouse. Molecules 2021, 26, 4203. [Google Scholar] [CrossRef]
  113. Hakimi, M.-A.; Olias, P.; Sibley, L.D. Toxoplasma Effectors Targeting Host Signaling and Transcription. Clin. Microbiol. Rev. 2017, 30, 615–645. [Google Scholar] [CrossRef] [Green Version]
  114. Ihara, F.; Nishikawa, Y. Toxoplasma gondii manipulates host cell signaling pathways via its secreted effector molecules. Parasitol. Int. 2021, 83, 102368. [Google Scholar] [CrossRef]
  115. Niedelman, W.; Gold, D.A.; Rosowski, E.; Sprokholt, J.K.; Lim, D.; Arenas, A.; Melo, M.; Spooner, E.; Yaffe, M.B.; Saeij, J.P.J. The rhoptry proteins ROP18 and ROP5 mediate Toxoplasma gondii evasion of the murine, but not the human, interferon-gamma response. PLoS Pathog. 2012, 8, e1002784. [Google Scholar] [CrossRef] [Green Version]
  116. El Hajj, H.; Demey, E.; Poncet, J.; Lebrun, M.; Wu, B.; Galéotti, N.; Fourmaux, M.N.; Mercereau-Puijalon, O.; Vial, H.; Labesse, G.; et al. The ROP2 family of Toxoplasma gondii rhoptry proteins: Proteomic and genomic characterization and molecular modeling. Proteomics 2006, 6, 5773–5784. [Google Scholar] [CrossRef]
  117. El Hajj, H.; Lebrun, M.; Arold, S.T.; Vial, H.; Labesse, G.; Dubremetz, J.F. ROP18 is a rhoptry kinase controlling the intracellular proliferation of Toxoplasma gondii. PLoS Pathog. 2007, 3, e14. [Google Scholar] [CrossRef] [Green Version]
  118. Sinai, A.P.; Joiner, K.A. The Toxoplasma gondii protein ROP2 mediates host organelle association with the parasitophorous vacuole membrane. J. Cell Biol. 2001, 154, 95–108. [Google Scholar] [CrossRef] [Green Version]
  119. Pernas, L.; Boothroyd, J.C. Association of host mitochondria with the parasitophorous vacuole during Toxoplasma infection is not dependent on rhoptry proteins ROP2/8. Int. J. Parasitol. 2010, 40, 1367–1371. [Google Scholar] [CrossRef] [Green Version]
  120. El Hajj, H.; Lebrun, M.; Fourmaux, M.N.; Vial, H.; Dubremetz, J.F. Inverted topology of the Toxoplasma gondii ROP5 rhoptry protein provides new insights into the association of the ROP2 protein family with the parasitophorous vacuole membrane. Cell. Microbiol. 2007, 9, 54–64. [Google Scholar] [CrossRef]
  121. Etheridge, R.D.; Alaganan, A.; Tang, K.; Lou, H.J.; Turk, B.E.; Sibley, L.D. The Toxoplasma pseudokinase ROP5 forms complexes with ROP18 and ROP17 kinases that synergize to control acute virulence in mice. Cell Host Microbe 2014, 15, 537–550. [Google Scholar] [CrossRef] [Green Version]
  122. Behnke, M.; Fentress, S.J.; Mashayekhi, M.; Li, L.X.; Taylor, G.A.; Sibley, L.D. The polymorphic pseudokinase ROP5 controls virulence in Toxoplasma gondii by regulating the active kinase ROP18. PLoS Pathog. 2012, 8, e1002992. [Google Scholar] [CrossRef]
  123. Bernstein, M.; Pardini, L.; Bello Pede Castro, B.; Unzaga, J.M.; Venturini, M.C.; More, G. ROP18 and ROP5 alleles combinations are related with virulence of T. gondii isolates from Argentina. Parasitol. Int. 2021, 83, 102328. [Google Scholar] [CrossRef]
  124. Shwab, E.K.; Jiang, T.; Pena, H.F.; Gennari, S.M.; Dubey, J.P.; Su, C. The ROP18 and ROP5 gene allele types are highly predictive of virulence in mice across globally distributed strains of Toxoplasma gondii. Int. J. Parasitol. 2016, 46, 141–146. [Google Scholar] [CrossRef]
  125. Rêgo, W.; Costa, J.; Baraviera, R.; Pinto, L.; Bessa, G.; Lopes, R.; Vitor, R. Association of ROP18 and ROP5 was efficient as a marker of virulence in atypical isolates of Toxoplasma gondii obtained from pigs and goats in Piaui, Brazil. Vet. Parasitol. 2017, 247, 19–25. [Google Scholar] [CrossRef]
  126. Wei, F.; Wang, W.; Liu, Q. Protein kinases of Toxoplasma gondii: Functions and drug targets. Parasitol. Res. 2013, 112, 2121–2129. [Google Scholar] [CrossRef]
  127. Grzybowski, M.M.; Dziadek, B.; Gatkowska, J.M.; Dzitko, K.; Dlugonska, H. Towards vaccine against toxoplasmosis: Evaluation of the immunogenic and protective activity of recombinant ROP5 and ROP18 Toxoplasma gondii proteins. Parasitol. Res. 2015, 114, 4553–4563. [Google Scholar] [CrossRef]
  128. Grzybowski, M.M.; Gatkowska, J.M.; Dziadek, B.; Dzitko, K.; Długońska, H. Human toxoplasmosis: A comparative evaluation of the diagnostic potential of recombinant Toxoplasma gondii ROP5 and ROP18 antigens. J. Med. Microbiol. 2015, 64, 1201–1207. [Google Scholar] [CrossRef]
  129. Behnke, M.; Khan, A.; Wootton, J.C.; Dubey, J.P.; Tang, K.; Sibley, L.D. Virulence differences in Toxoplasma mediated by amplification of a family of polymorphic pseudokinases. Proc. Natl. Acad. Sci. USA 2011, 108, 9631–9636. [Google Scholar] [CrossRef] [Green Version]
  130. Blader, I.J.; Saeij, J.P. Communication between Toxoplasma gondii and its host: Impact on parasite growth, development, immune evasion, and virulence. APMIS 2009, 117, 458–476. [Google Scholar] [CrossRef] [Green Version]
  131. Taylor, S.; Barragan, A.; Su, C.; Fux, B.; Fentress, S.J.; Tang, K.; Beatty, W.L.; El Hajj, H.; Jerome, M.; Behnke, M.S.; et al. A secreted serine-threonine kinase determines virulence in the eukaryotic pathogen Toxoplasma gondii. Science 2006, 314, 1776–1780. [Google Scholar] [CrossRef] [Green Version]
  132. Fentress, S.J.; Behnke, M.S.; Dunay, I.R.; Mashayekhi, M.; Rommereim, L.M.; Fox, B.A.; Bzik, D.J.; Taylor, G.A.; Turk, B.E.; Lichti, C.F.; et al. Phosphorylation of immunity-related GTPases by a Toxoplasma gondii-secreted kinase promotes macrophage survival and virulence. Cell Host Microbe 2010, 8, 484–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Steinfeldt, T.; Konen-Waisman, S.; Tong, L.; Pawlowski, N.; Lamkemeyer, T.; Sibley, L.D.; Hunn, J.P.; Howard, J.C. Phosphorylation of mouse immunity-related GTPase (IRG) resistance proteins is an evasion strategy for virulent Toxoplasma gondii. PLoS Biol. 2010, 8, e1000576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Butcher, B.A.; Fox, B.A.; Rommereim, L.M.; Kim, S.G.; Maurer, K.J.; Yarovinsky, F.; Herbert, D.B.R.; Bzik, D.J.; Denkers, E.Y. Toxoplasma gondii rhoptry kinase ROP16 activates STAT3 and STAT6 resulting in cytokine inhibition and arginase-1-dependent growth control. PLoS Pathog. 2011, 7, e1002236. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, L.; Christian, D.A.; Kochanowsky, J.A.; Phan, A.T.; Clark, J.T.; Wang, S.; Berry, C.; Oh, J.; Chen, X.; Roos, D.S.; et al. The Toxoplasma gondii virulence factor ROP16 acts in cis and trans, and suppresses T cell responses. J. Exp. Med. 2020, 217, e20181757. [Google Scholar] [CrossRef]
  136. Kochanowsky, J.A.; Thomas, K.K.; Koshy, A.A. ROP16-Mediated Activation of STAT6 Suppresses Host Cell Reactive Oxygen Species Production, Facilitating Type III Toxoplasma gondii Growth and Survival. mBio 2021, 12, e03305-20. [Google Scholar] [CrossRef]
  137. Sabou, M.; Doderer-Lang, C.; Leyer, C.; Konjic, A.; Kubina, S.; Lennon, S.; Rohr, O.; Viville, S.; Cianférani, S.; Candolfi, E.; et al. Toxoplasma gondii ROP16 kinase silences the cyclin B1 gene promoter by hijacking host cell UHRF1-dependent epigenetic pathways. Cell. Mol. Life Sci. 2020, 77, 2141–2156. [Google Scholar] [CrossRef] [Green Version]
  138. Simpson, C.; Jones, N.G.; Hull-Ryde, E.A.; Kireev, D.; Stashko, M.; Tang, K.; Janetka, J.W.; Wildman, S.A.; Zuercher, W.J.; Schapira, M.; et al. Identification of small molecule inhibitors that block the Toxoplasma gondii rhoptry kinase ROP18. ACS Infect. Dis. 2016, 2, 194–206. [Google Scholar] [CrossRef] [Green Version]
  139. Molina, D.; Cossio-Pérez, R.; Rocha-Roa, C.; Pedraza, L.; Cortes, E.; Hernández, A.; Gómez-Marín, J.E. Protein targets of thiazolidinone derivatives in Toxoplasma gondii and insights into their binding to ROP18. BMC Genom. 2018, 19, 856. [Google Scholar] [CrossRef]
  140. Maclean, A.E.; Bridges, H.R.; Silva, M.F.; Ding, S.; Ovciarikova, J.; Hirst, J.; Sheiner, L. Complexome profile of Toxoplasma gondii mitochondria identifies divergent subunits of respiratory chain complexes including new subunits of cytochrome bc1 complex. PLoS Pathog. 2021, 17, e1009301. [Google Scholar] [CrossRef]
  141. Al-Anouti, F.; Tomavo, S.; Parmley, S.; Ananvoranich, S. The expression of lactate dehydrogenase is important for the cell cycle of Toxoplasma gondii. J. Biol. Chem. 2004, 279, 52300–52311. [Google Scholar] [CrossRef] [Green Version]
  142. Alday, P.H.; Bruzual, I.; Nilsen, A.; Pou, S.; Winter, R.; Ben Mamoun, C.; Riscoe, M.K.; Doggett, J.S. Genetic Evidence for Cytochrome b Qi Site Inhibition by 4(1H)-Quinolone-3-Diarylethers and Antimycin in Toxoplasma gondii. Antimicrob. Agents Chemother. 2017, 61, e01866-16. [Google Scholar] [CrossRef] [Green Version]
  143. McConnell, E.V.; Bruzual, I.; Pou, S.; Winter, R.; Dodean, R.A.; Smilkstein, M.J.; Krollenbrock, A.; Nilsen, A.; Zakharov, L.N.; Riscoe, M.K.; et al. Targeted Structure-Activity Analysis of Endochin-like Quinolones Reveals Potent Qi and Qo Site Inhibitors of Toxoplasma gondii and Plasmodium falciparum Cytochrome bc1 and Identifies ELQ-400 as a Remarkably Effective Compound against Acute Experimental Toxoplasmosis. ACS Infect. Dis. 2018, 4, 1574–1584. [Google Scholar]
  144. Doggett, J.S.; Nilsen, A.; Forquer, I.; Wegmann, K.W.; Jones-Brando, L.; Yolken, R.H.; Bordón, C.; Charman, S.A.; Katneni, K.; Schultz, T.; et al. Endochin-like quinolones are highly efficacious against acute and latent experimental toxoplasmosis. Proc. Natl. Acad. Sci. USA 2012, 109, 15936–15941. [Google Scholar] [CrossRef] [Green Version]
  145. Secrieru, A.; Costa, I.C.C.; O’Neill, P.M.; Cristiano, M.L.S. Antimalarial Agents as Therapeutic Tools against Toxoplasmosis—A Short Bridge. between Two Distant Illnesses. Molecules 2020, 25, 1574. [Google Scholar] [CrossRef] [Green Version]
  146. Bougdour, A.; Maubon, D.; Baldacci, P.; Ortet, P.; Bastien, O.; Bouillon, A.; Barale, J.-C.; Pelloux, H.; Ménard, R.; Hakimi, M.-A. Drug inhibition of HDAC3 and epigenetic control of differentiation in Apicomplexa parasites. J. Exp. Med. 2009, 206, 953–966. [Google Scholar] [CrossRef]
  147. Maubon, D.; Bougdour, A.; Wong, Y.-S.; Brenier-Pinchart, M.-P.; Curt, A.; Hakimi, M.-A.; Pelloux, H. 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. 2010, 54, 4843–4850. [Google Scholar] [CrossRef] [Green Version]
  148. Afifi, M.A.; Al-Rabia, M.W. The immunomodulatory effects of rolipram abolish drug-resistant latent phase of Toxoplasma gondii infection in a murine model. J. Microsc. Ultrastruct. 2015, 3, 86–91. [Google Scholar] [CrossRef]
  149. Wei, S.; Marches, F.; Daniel, B.; Sonda, S.; Heidenreich, K.; Curiel, T. Pyridinylimidazole p38 mitogen-activated protein kinase inhibitors block intracellular Toxoplasma gondii replication. Int. J. Parasitol. 2002, 32, 969–977. [Google Scholar] [CrossRef]
  150. Brumlik, M.J.; Pandeswara, S.; Ludwig, S.M.; Jeansonne, D.P.; Lacey, M.R.; Murthy, K.; Daniel, B.J.; Wang, R.F.; Thibodeaux, S.R.; Church, K.M.; et al. TgMAPK1 is a Toxoplasma gondii MAP kinase that hijacks host MKK3 signals to regulate virulence and interferon-gamma-mediated nitric oxide production. Exp. Parasitol. 2013, 134, 389–399. [Google Scholar] [CrossRef] [Green Version]
  151. Brumlik, M.J.; Wei, S.; Finstad, K.; Nesbit, J.; Hyman, L.E.; Lacey, M.; Burow, M.E.; Curiel, T.J. Identification of a novel mitogen-activated protein kinase in Toxoplasma gondii. Int. J. Parasitol. 2004, 34, 1245–1254. [Google Scholar] [CrossRef]
  152. Sun, H.; Zhuo, X.; Zhao, X.; Yang, Y.; Chen, X.; Yao, C.; Du, A. The heat shock protein 90 of Toxoplasma gondii is essential for invasion of host cells and tachyzoite growth. Parasite 2017, 24, 22. [Google Scholar] [CrossRef] [Green Version]
  153. Lyons, R.E.; Johnson, A.M. Heat shock proteins of Toxoplasma gondii. Parasite Immunol. 1995, 17, 353–359. [Google Scholar] [CrossRef]
  154. Toursel, C.; Dzierszinski, F.; Bernigaud, A.; Mortuaire, M.; Tomavo, S. Molecular cloning, organellar targeting and developmental expression of mitochondrial chaperone HSP60 in Toxoplasma gondii. Mol. Biochem. Parasitol. 2000, 111, 319–332. [Google Scholar] [CrossRef]
  155. Dobbin, C.A.; Smith, N.C.; Johnson, A.M. Heat shock protein 70 is a potential virulence factor in murine toxoplasma infection via immunomodulation of host NF-kappa B and nitric oxide. J. Immunol. 2002, 169, 958–965. [Google Scholar] [CrossRef] [Green Version]
  156. Ashwinder, K.; Kho, M.T.; Chee, P.M.; Lim, W.Z.; Yap, I.K.S.; Choi, S.B.; Yam, W.K. Targeting Heat Shock Proteins 60 and 70 of Toxoplasma gondii as a Potential Drug Target.: In Silico Approach. Interdiscip. Sci. 2016, 8, 374–387. [Google Scholar] [CrossRef]
  157. Boyom, F.F.; Fokou, P.V.T.; Tchokouaha, L.R.Y.; Spangenberg, T.; Mfopa, A.N.; Kouipou, R.M.T.; Mbouna, C.J.; Donfack, V.F.D.; Zollo, P.H.A. Repurposing the open access malaria box to discover potent inhibitors of Toxoplasma gondii and Entamoeba histolytica. Antimicrob. Agents Chemother. 2014, 58, 5848–5854. [Google Scholar] [CrossRef] [Green Version]
  158. Spalenka, J.; Escotte-Binet, S.; Bakiri, A.; Hubert, J.; Renault, J.-H.; Velard, F.; Duchateau, S.; Aubert, D.; Huguenin, A.; Villena, I. Discovery of New Inhibitors of Toxoplasma gondii via the Pathogen Box. Antimicrob. Agents Chemother. 2018, 62, e01640-17. [Google Scholar] [CrossRef] [Green Version]
  159. Murata, Y.; Sugi, T.; Weiss, L.M.; Kato, K. Identification of compounds that suppress Toxoplasma gondii tachyzoites and bradyzoites. PLoS ONE 2017, 12, e0178203. [Google Scholar] [CrossRef] [Green Version]
  160. Opsenica, I.; Verbić, T.; Tot, M.; Sciotti, R.; Pybus, B.S.; Djurković-Djaković, O.; Slavic, K.; Šolaja, B.A. Investigation into novel thiophene- and furan-based 4-amino-7-chloroquinolines afforded antimalarials that cure mice. Bioorg. Med. Chem. 2015, 23, 2176–2186. [Google Scholar] [CrossRef]
  161. Eissa, M.M.; Barakat, A.M.; Amer, E.I.; Younis, L.K. Could miltefosine be used as a therapy for toxoplasmosis? Exp. Parasitol. 2015, 157, 12–22. [Google Scholar] [CrossRef]
  162. Liu, S.; Wu, M.; Hua, Q.; Lu, D.; Tian, Y.; Yu, H.; Cheng, L.; Chen, Y.; Cao, J.; Hu, X.; et al. Two old drugs, NVP-AEW541 and GSK-J4, repurposed against the Toxoplasma gondii RH strain. Parasites Vectors 2020, 13, 242. [Google Scholar] [CrossRef] [PubMed]
  163. Dittmar, A.J.; Drozda, A.A.; Blader, I.J. Drug Repurposing Screening Identifies Novel Compounds That Effectively Inhibit Toxoplasma gondii Growth. mSphere 2016, 1, e00042-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Angel, S.O.; Vanagas, L.; Ruiz, D.M.; Cristaldi, C.; Cartagena, A.M.S.; Sullivan, W.J.J. Emerging Therapeutic Targets Against Toxoplasma gondii: Update on DNA Repair Response Inhibitors and Genotoxic Drugs. Front. Cell. Infect. Microbiol. 2020, 10, 289. [Google Scholar] [CrossRef] [PubMed]
  165. Adeyemi, O.; Atolani, O.; Awakan, O.J.; Olaolu, T.D.; Nwonuma, C.O.; Alejolowo, O.; Otohinoyi, D.A.; Rotimi, D.; Owolabi, A.; Batiha, G. In Vitro Screening to Identify Anti-Toxoplasma Compounds and In Silico Modeling for Bioactivities and Toxicity. Yale J. Biol. Med. 2019, 92, 369–383. [Google Scholar]
  166. Barbosa, B.F.; Gomes, A.O.; Ferro, E.; Napolitano, D.R.; Mineo, J.R.; Silva, N.M. Enrofloxacin is able to control Toxoplasma gondii infection in both in vitro and in vivo experimental models. Vet. Parasitol. 2012, 187, 44–52. [Google Scholar] [CrossRef]
  167. Khan, A.A.; Slifer, T.R.; Araujo, F.G.; Remington, J.S. Activity of gatifloxacin alone or in combination with Pyrimethamine or gamma interferon against Toxoplasma gondii. Antimicrob. Agents Chemother. 2001, 45, 48–51. [Google Scholar] [CrossRef] [Green Version]
  168. Jones-Brando, L.; D’Angelo, J.; Posner, G.H.; Yolken, R. In vitro inhibition of Toxoplasma gondii by four new derivatives of artemisinin. Antimicrob. Agents Chemother. 2006, 50, 4206–4208. [Google Scholar] [CrossRef] [Green Version]
  169. Dunay, I.R.; Chan, W.C.; Haynes, R.; Sibley, L.D. Artemisone and artemiside control acute and reactivated toxoplasmosis in a murine model. Antimicrob. Agents Chemother. 2009, 53, 4450–4456. [Google Scholar] [CrossRef] [Green Version]
  170. D’Angelo, J.G.; Bordón, C.; Posner, G.H.; Yolken, R.; Jones-Brando, L. Artemisinin derivatives inhibit Toxoplasma gondii in vitro at multiple steps in the lytic cycle. J. Antimicrob. Chemother. 2009, 63, 146–150. [Google Scholar] [CrossRef] [Green Version]
  171. Schultz, T.L.; Hencken, C.P.; Woodard, L.E.; Posner, G.H.; Yolken, R.H.; Jones-Brando, L.; Carruthers, V.B. A thiazole derivative of artemisinin moderately reduces Toxoplasma gondii cyst burden in infected mice. J. Parasitol. 2014, 100, 516–521. [Google Scholar] [CrossRef]
  172. Alomar, M.L.; Rasse-Suriani, F.A.; Ganuza, A.; Coceres, V.M.; Cabrerizo, F.M.; Angel, S.O. In vitro evaluation of beta-carboline alkaloids as potential anti-Toxoplasma agents. BMC Res. Notes 2013, 6, 193. [Google Scholar] [CrossRef] [Green Version]
  173. Goodwin, D.G.; Strobl, J.S.; Lindsay, D.S. Evaluation of five antischizophrenic agents against Toxoplasma gondii in human cell cultures. J. Parasitol. 2011, 97, 148–151. [Google Scholar] [CrossRef] [Green Version]
  174. Messina, M.; Niesman, I.; Mercier, C.; Sibley, L. Stable DNA transformation of Toxoplasma gondii using phleomycin selection. Gene 1995, 165, 213–217. [Google Scholar] [CrossRef]
  175. Tenorio, R.P.; Carvalho, C.S.; Pessanha, C.S.; de Lima, J.G.; de Faria, A.R.; Alves, A.J.; de Melo, E.J.; Goes, A.J. Synthesis of thiosemicarbazone and 4-thiazolidinone derivatives and their in vitro anti-Toxoplasma gondii activity. Bioorg. Med. Chem. Lett. 2005, 15, 2575–2578. [Google Scholar] [CrossRef]
  176. Jones-Brando, L.; Torrey, E.F.; Yolken, R. Drugs used in the treatment of schizophrenia and bipolar disorder inhibit the replication of Toxoplasma gondii. Schizophr. Res. 2003, 62, 237–244. [Google Scholar] [CrossRef]
  177. Miller, R.L.; Gerster, J.F.; Owens, M.L.; Slade, H.B.; Tomai, M.A. Imiquimod applied topically: A novel immune response modifier and new class of drug. Int. J. Immunopharmacol. 1999, 21, 1–14. [Google Scholar] [CrossRef]
  178. Miranda-Verastegui, C.; Tulliano, G.; Gyorkos, T.W.; Calderon, W.; Rahme, E.; Ward, B.; Cruz, M.; Llanos-Cuentas, A.; Matlashewski, G. First-line therapy for human cutaneous leishmaniasis in Peru using the TLR7 agonist imiquimod in combination with pentavalent antimony. PLoS Negl. Trop. Dis. 2009, 3, e491. [Google Scholar] [CrossRef]
  179. Raman, V.S.; Duthie, M.S.; Fox, C.B.; Matlashewski, G.; Reed, S.G. Adjuvants for Leishmania vaccines: From models to clinical application. Front. Immunol. 2012, 3, 144. [Google Scholar] [CrossRef] [Green Version]
  180. El Hajj, R.; Youness, H.B.; Lachaud, L.; Bastien, P.; Masquefa, C.; Bonnet, P.-A.; El Hajj, H.; Khalifeh, I. EAPB0503: An Imiquimod analog with potent in vitro activity against cutaneous leishmaniasis caused by Leishmania major and Leishmania tropica. PLoS Negl. Trop. Dis. 2018, 12, e0006854. [Google Scholar] [CrossRef] [Green Version]
  181. Miranda-Verastegui, C.; Llanos-Cuentas, A.; Arevalo, I.; Ward, B.J.; Matlashewski, G. Randomized, double-blind clinical trial of topical imiquimod 5% with parenteral meglumine antimoniate in the treatment of cutaneous leishmaniasis in Peru. Clin. Infect. Dis. 2005, 40, 1395–1403. [Google Scholar] [CrossRef]
  182. Hamie, M.; Najm, R.; Deleuze-Masquefa, C.; Bonnet, P.A.; Dubremetz, J.F.; El Sabban, M.; El Hajj, H. Imiquimod Targets Toxoplasmosis Through Modulating Host Toll-Like Receptor-MyD88 Signaling. Front. Immunol. 2021, 12, 629917. [Google Scholar] [CrossRef]
  183. Sun, Q.; He, M.; Zhang, M.; Zeng, S.; Chen, L.; Zhou, L.; Xu, H. Ursolic acid: A systematic review of its pharmacology, toxicity and rethink on its pharmacokinetics based on PK-PD model. Fitoterapia 2020, 147, 104735. [Google Scholar] [CrossRef]
  184. Choi, W.H.; Lee, I.A. The Mechanism of Action of Ursolic Acid as a Potential Anti-Toxoplasmosis Agent, and Its Immunomodulatory Effects. Pathogens 2019, 8, 61. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation of known Toxoplasma gondii therapeutic targets.
Figure 1. Schematic representation of known Toxoplasma gondii therapeutic targets.
Microorganisms 09 02531 g001
Table 1. Therapeutic modalities of toxoplasmosis.
Table 1. Therapeutic modalities of toxoplasmosis.
ToxoplasmosisCurrently Used DrugsMechanism(s) of Action
Congenital toxoplasmosis
Maternal congenital toxoplasmosis or confirmed infection of neonate or fetus following congenital toxoplasmosis1 Pyrimethamine (Inhibitor of dihydrofolate reductase (DHFR))Inhibition of the biosynthesis of parasitic folate, interrupting nucleic acid synthesis and parasite replication
+ 1 Sulfadiazine (Inhibitor of dihydropteroate synthase)
+ Folinic acid (leucovorin)Reduction of the harmful side effects (i.e., bone marrow myelosuppression)
Suspected congenital toxoplasmosisSpiramycinInhibition of protein synthesis
Toxoplasmosis in immunocompetent patients
Acute toxoplasmosis2 Pyrimethamine (Inhibitor of dihydrofolate reductase (DHFR))Inhibition of the biosynthesis of parasitic folate, interrupting nucleic acid synthesis and parasite replication
+ 2 Sulfadiazine (Inhibitor of dihydropteroate synthase)
+ Folinic acid (leucovorin)
Reduction of the harmful side effects (i.e., bone marrow myelosuppression)
2 PyrimethamineInhibition of the biosynthesis of parasitic folate
+3 ClindamycinInhibition of protein synthesis
+ Folinic acidReduction of the harmful side effects (i.e., bone marrow myelosuppression)
2 PyrimethamineInhibition of the biosynthesis of parasitic folate
+ Folinic acidReduction of the harmful side effects (i.e., bone marrow myelosuppression)
+ AtovaquoneTargeting the mitochondrial electron transport and the mitochondrial cytochrome bc1 complex
2 PyrimethamineInhibition of the biosynthesis of parasitic folate
Reduction of the harmful side effects (i.e., bone marrow myelosuppression)
+ Folinic acid
Inhibition of protein synthesis
+ 4 Azithromycin
2 TrimethoprimInhibition of the biosynthesis of parasitic folate
+ SulfamethoxazoleBacteriostatic sulfonamide interfering with folic acid synthesis
Ocular toxoplasmosis
PyrimethamineInhibition of the biosynthesis of parasitic folate, interrupting nucleic acid synthesis and parasite replication
+ Sulfadiazine
+/− 5 Steroids
Intravitreal ClindamycinInhibition of protein synthesis
+ 5 Steroids
TrimethoprimInhibition of the biosynthesis of parasitic folate
+ SulfamethoxazoleBacteriostatic sulfonamide interfering with folic acid synthesis
+ 2 Steroids
AtovaquoneTargeting the mitochondrial electron transport and the mitochondrial cytochrome bc1 complex
4 Azitromycin+/− PyrimethamineInhibition of protein synthesis+/− biosynthesis of parasite folate
Toxoplasmosis in immunocompromised patients
(cycles of different doses in induction and maintenance therapy)
2 PyrimethamineInhibition of the biosynthesis of parasitic folate, interrupting nucleic acid synthesis and parasite replication
+ 2 SulfadiazineReduction of the harmful side effects
+ Folinic acid (leucovorin)
2 PyrimethamineInhibition of the biosynthesis of parasitic folate
+ 3 ClindamycinInhibition of protein synthesis
+ Folinic acidReduction of the harmful side effects
2 TrimethoprimInhibition of the biosynthesis of parasitic folate
+ SulfamethoxazoleBacteriostatic sulfonamide interfering with folic acid synthesis
2 PyrimethamineInhibition of the biosynthesis of parasitic folate
+ Folinic acidReduction of the harmful side effects (i.e., bone marrow myelosuppression)
Targeting the mitochondrial electron transport and the mitochondrial cytochrome bc1 complex
+ Atovaquone
2 SulfadiazineInhibition of the biosynthesis of parasitic folate
+ AtovaquoneTargeting the mitochondrial electron transport and the mitochondrial cytochrome bc1 complex
2 PyrimethamineInhibition of the biosynthesis of parasitic folate
+ Folinic acidReduction of the harmful side effects (i.e., bone marrow myelosuppression)
Inhibition of protein synthesis
+ 4 Azithromycin
1 Side effects: Hematologic side effects (neutropenia, thrombocytopenia, and leukopenia, among others), in rare cases agranulocytosis, toxic epidermal necrolysis, hepatic necrosis, teratogenic potential if used during the first trimester of pregnancy. 2 Side effects: Hematologic side effects (neutropenia, thrombocytopenia, and leukopenia, among others), elevated liver enzymes, elevated creatinine levels. 3 Side effects: may cause diarrhea, and Clostridium difficile infection. 4 Side effects: may be associated with hearing problems. 5 Side effects: Steroids can have detrimental effects on vision (endophthalmitis) and vision loss if used without concomitant antimicrobial therapy.
Table 2. Summary of Toxoplasma gondii therapeutic targets and their corresponding drugs.
Table 2. Summary of Toxoplasma gondii therapeutic targets and their corresponding drugs.
Parasite Targets
ApicoplastMicronemesRhoptriesMitochondriaNucleus
Inhibitors of Fatty
Acid synthesis:
-Clodinafop
-Thiolactomycin
-Triclosan
BKI targeting TgCDPK1:
-BKI-1294
-BKI-1294 analogs:
Compounds 24 and 32)
-BKI-1748
OxindolesTargeting HSP60Topo-isomerase 2 inhibitors:
-Daunorubicin
-Trovafloxacin
-Enrofloxacin
-Gatofloxacin
Inhibitors of 2-Isoprenoid synthesis:
-Fosmidomycin
SP2306-azaquinazolinesAtovaquoneTopo-isomerase 1 inhibitors:
-Artemisinin
-Artemisone
-Artemiside
-Artemether
-Harmane
-Harmine
-Non-harmane
Inhibitors of DNA gyrase:
-Quinolones
-Fuoroquinolones
-Ciprofloxacin
-Trovafloxacin
-Ofloxacin
-Temafloxacin
PyrazolopyridinesELQ-271DNA-intercalating agents:
-Fluphenasine
-Thioridazine
-Trifluoperazine
-Hycanton
-Phleomycin
-Mitomycin C
Inhibitors of Protein synthesis:
-Clindamycin
-Spiramycin
-Azithromycin
Chemical scaffoldsELQ-316Ribonucleotide reductase inhibitors:
-Thiosemicarbazones
-Hydroxyurea
Thiazolidinone derivativesELQ-400Oxidative DNA damage/DNA binding:
-Resveratrol
-Valproic acid
Naphtoquinones
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hajj, R.E.; Tawk, L.; Itani, S.; Hamie, M.; Ezzeddine, J.; El Sabban, M.; El Hajj, H. Toxoplasmosis: Current and Emerging Parasite Druggable Targets. Microorganisms 2021, 9, 2531. https://doi.org/10.3390/microorganisms9122531

AMA Style

Hajj RE, Tawk L, Itani S, Hamie M, Ezzeddine J, El Sabban M, El Hajj H. Toxoplasmosis: Current and Emerging Parasite Druggable Targets. Microorganisms. 2021; 9(12):2531. https://doi.org/10.3390/microorganisms9122531

Chicago/Turabian Style

Hajj, Rana El, Lina Tawk, Shaymaa Itani, Maguy Hamie, Jana Ezzeddine, Marwan El Sabban, and Hiba El Hajj. 2021. "Toxoplasmosis: Current and Emerging Parasite Druggable Targets" Microorganisms 9, no. 12: 2531. https://doi.org/10.3390/microorganisms9122531

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop