Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 81

Warning: fopen(upload/ip_log/ip_log_2024-04.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 83

Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 84
Recent Advances in Toxoplasma gondii Immunotherapeutics
| Home | E-Submission | Sitemap | Contact us |  
top_img
Korean J Parasito Search

CLOSE

Korean J Parasito > Volume 52(6):2014 > Article
Lim and Othman: Recent Advances in Toxoplasma gondii Immunotherapeutics

Abstract

Toxoplasmosis is an opportunistic infection caused by the protozoan parasite Toxoplasma gondii. T. gondii is widespread globally and causes severe diseases in individuals with impaired immune defences as well as congenitally infected infants. The high prevalence rate in some parts of the world such as South America and Africa, coupled with the current drug treatments that trigger hypersensitivity reactions, makes the development of immunotherapeutics intervention a highly important research priority. Immunotherapeutics strategies could either be a vaccine which would confer a pre-emptive immunity to infection, or passive immunization in cases of disease recrudescence or recurrent clinical diseases. As the severity of clinical manifestations is often greater in developing nations, the development of well-tolerated and safe immunotherapeutics becomes not only a scientific pursuit, but a humanitarian enterprise. In the last few years, much progress has been made in vaccine research with new antigens, novel adjuvants, and innovative vaccine delivery such as nanoparticles and antigen encapsulations. A literature search over the past 5 years showed that most experimental studies were focused on DNA vaccination at 52%, followed by protein vaccination which formed 36% of the studies, live attenuated vaccinations at 9%, and heterologous vaccination at 3%; while there were few on passive immunization. Recent progress in studies on vaccination, passive immunization, as well as insights gained from these immunotherapeutics is highlighted in this review.

INTRODUCTION

Toxoplasmosis is a zoonotic disease caused by the obligate intracellular parasite Toxoplasma gondii. Toxoplasmosis can cause severe neurologic, ocular, and systemic diseases in neonates and individuals with weakened immune system [1]. In immunocompetent adults, T. gondii infection is usually asymptomatic and forms dormant tissue cysts containing bradyzoites at immune-privileged sites such as the brain and muscles [1]. When immunity is impaired, latent infection is then reactivated causing widespread tissue destruction and severe pathology, and is among the major causes of mortality in AIDS patients. Disease severity is exacerbated in underdeveloped countries with limited financial resources and infrastructure to acquire and distribute the anti-retroviral drugs [2]. The sub-Saharan African continent is a dire example of this. A high rate of HIV infection combined with high seroprevalence of T. gondii co-infection would result in an estimated 2.5 to 10 million people in this region who are at risk of dying from toxoplasmic encephalitis [3]. Atypical strains, predominantly found in South and Central America, have caused debilitating diseases, especially in the form of recurrent ocular disease that can lead to blindness in otherwise healthy adults [4,5]. This is an especially critical problem in Brazil where the current risk of ocular toxoplasmosis is at a high rate of 18% [6]. Thus, this pathogen remains as a severe public health problem in developing countries.
The development of a vaccine against toxoplasmosis is highly important to arrest the spread of the disease because in theory, a single treatment confers life-long protective immunity. Significant strides have been made in the past 15 years in antigen isolation and characterization, gene-cloning, generation of parasite mutants, and immunological methods [7]. Vaccine studies demonstrating robust disease control are antigens that could elicit a protective Th1-biased immune response and generation of long-lived cytotoxic CD8+ T cells producing IFN-γ [8]. Vaccinations have thus far proven effective at reducing disease mortality and parasite cyst burden in mice, but do not achieve complete protection. Thus, sterile immunity from the disease remains an elusive target.
As toxoplasmosis is an opportunistic infection causing illness in immunocompromised patients with reduced cellular immunity, therapeutic intervention to reduce pathogenesis is also needed. Current anti-parasitic drug treatments cause toxic hypersensitivity reactions and are teratogenic, presenting a need for safer and well-tolerated treatment options. As the major cause of toxoplasmosis mortality is due to reactivation of latent infection, while considerable morbidity can result from the tendency of congenital infections to relapse with retinal damage, studies on passive immunization to decrease parasitemia burden is necessary for limiting the disease pathology. The aim of this review is to focus on recent advances in both the active and passive immunization studies against toxoplasmosis in murine models.

VACCINES

Based on observations that chronic infection is able to confer immunological memory to pregnant mothers, normally preventing disease transmission to fetuses even if re-exposed during gestation, a toxoplasmosis vaccine seems highly feasible. Various vaccination strategies against toxoplasmosis in rodent models have been employed in the past decades, ranging from inactivated vaccines, protein vaccines in subunit or multi-antigenic cocktails, and DNA vaccines. However, efficacious vaccines remains a challenge as vaccine experiments have not been able to confer sterile immunity against infection. Sterile immunity is a protection endpoint that is assessed not only by survival rate, but also by the lack of tissue cysts after challenge infection, which is a hallmark of chronic infection. Only a few live, attenuated strains of T. gondii have been able to elicit a potent Th1 immune response for effective disease resistance, but live vaccines such as the commercially-licensed ToxoVax® (Intervet B.V.) used for sheep immunization is considered unsafe for human use and is restricted in its applications due to safety concerns as well as a short shelf-life.
It has been established that the IFN-γ-secreting CD8+ T lymphocyte is the major T cell subset involved in crucial long-term protective immunity in toxoplasmosis [9,10]. Thus, an ideal vaccine to treat toxoplasmosis should be sufficiently immunogenic to induce a Th1-type immune response to control disease pathogenesis, as well as to prevent the development of chronic tissue cysts. There are several challenging features in toxoplasmosis vaccine development. First is the stage-specific expression of parasite proteins which may render vaccines targeted to the acute stage ineffective at the chronic stage and vice versa. Second is the presence of varied T. gondii genotype strains means that vaccinations against a particular strain may not be protective against infections with other strains, especially with growing concern over the virulent, atypical strains from South America [11]. Third is the parasite's innate immune evasion mechanisms that enable the sequestration of dormant tissue cysts as a life-long chronic infection has been recalcitrant to both drug treatments and vaccination attempts to eliminate infection.

PROTEIN VACCINES

Significant advances made in the past decade in understanding the cell and molecular biology of the T. gondii parasite has provided the impetus to the current focus of vaccines based on defined subcellular components of the parasite. Among the parasite antigens that have been researched extensively in vaccination experiments are the surface antigen glycoproteins, or SAGs. Notably, investigations on highly immunogenic SAG1 expressed on tachyzoites have repeatedly yielded high survival rates and significant reduction in brain tissue cysts load [reviewed in 7]. SAG1 immunization studies by Khan et al. [12] and Debard et al. [13] demonstrated 100% and 85% reduction in brain cysts load when adjuvanted with QuilA and cholera toxin (CT) respectively. In a move to use less toxic adjuvants, a SAG1 vaccine co-administered with non-toxic heat-labile enterotoxin (LT) also provided significant but slightly lowered protection and a 78% reduction in brain cyst loads following intranasal delivery [14]. However, this highlights the need to develop adjuvants to simultaneously maximize immunostimulatory efficacy and safety. A recent development was the use of saponins from Panax ginseng roots as a non-toxic adjuvant in a ROP18 vaccine formulation, which improved protective efficacy of the vaccine [15]. Thus, these ginseng roots that have been used as traditional medicine hold potential in enhancement of the anti-toxoplasmosis immune responses and could be investigated further in vaccine formulations.
Immunity against T. gondii is highly complex and studies have shown how differences in host genetic background can influence vastly divergent disease outcome. This was apparent in a SAG1 vaccination which proved inhibitory to the materno-fetal transmission of toxoplasmosis in BALB/c mice by 50%, but not with CBA/J mice, which in fact exhibited increased disease transmission to fetuses [16]. As congenital toxoplasmosis can cause severe damage to a baby's brain, eyes, and nervous system, and current drug treatments are teratogenic, immunization efficacy in limiting congenital transmission should be also considered an important endpoint in more vaccine research.
In recent years, vaccination with several protein antigen combinations resulted in increased survival rates in mice following lethal challenge with type I parasite. The most significant gains in survival rates were reported for vaccine antigen SAG1 (80%) [17], SAG1+SAG2 (83%) [18], TgACT (50%) [19], and TgRACK-1 (45%) [20]. Among these findings, the unique study by Chuang et al. [17] used poly (lactide-co-glycolide) (PLG) microparticle encapsulation of the SAG1 and SAG2 protein antigens as a sustained-release vaccine formulation, which is an attractive concept for inducing prolonged disease immunity. Overall, with proper optimization to prevent antigens from denaturing during the encapsulation process, this is a strategy which could be also applied in various other immunotherapeutics formats to maintain therapeutic levels in blood or tissues for extended periods of time.
An important endpoint in the evaluation of toxoplasmosis vaccines is the provision of sterile immunity from the cystic bradyzoite stage, which is recalcitrant to current drug treatments and the source of disease reactivation in chronically-infected individuals. Thus, scoring the effects of immunization on brain tissue cysts loads in mice model gives crucial insights on vaccine utility. In lieu of this, the parasite antigens that have been included in numerous multi-antigen vaccines with good success are the ROP2 and GRA4 proteins. ROP2 is an abundant rhoptry protein essential in parasite multiplication and host invasion [21]. GRA4 is a dense granule protein localized to the intravacuolar network of the parasitophorous vacuole (PV) and involved in nutrient acquisition [22]. Both antigens have shown Th1-type immunogenicity, and its protective efficacy in various mouse strains [23] indicates these antigens as vaccine candidates that warrant further studies. The highest reductions in brain tissue cyst loads conferred were through the immunization with protein antigen combinations of ROP2+ROP4+SAG1 (90%) [24], ROP2+ROP4+GRA4 (84%) [25], and GRA7 nanoparticles (72%) [26]. In particular, the GRA7 nanoparticle is a novel vaccine based on monomeric T cell epitopes of GRA7 protein that self assembles into nanoparticles similar to a viral capsid. This nanoparticle vaccine is self-adjuvanting and demonstrates partial protective immunity to both type I and type II challenge infections.
Another interesting development was the vaccination with a recombinant plant-produced GRA4 antigen [27]. While a plant-produced toxoplasmosis antigen is not novel in itself [28], this work done by del L. Yácono et al. [27] involved the oral immunization of each mouse with leaf extracts equivalent to approximately 0.5 µg of GRA4 antigen. The substantial reduction in tissue cysts (59%) achieved by a low-dose vaccine followed by challenge with the cyst-forming type II strain potentiates the GRA4 antigen as a strong vaccine candidate. As T. gondii parasites mainly gain host entry by the intestinal mucosa route, an oral vaccine that can be potentially produced at lowered cost in plants shows good prospects. A summary of some of the most recent studies in protein vaccines against toxoplasmosis is provided in Table 1.

DNA VACCINES

DNA vaccination has the distinct advantage of being capable of generating an effective immune response with substantial cost benefits. DNA vaccinations against toxoplasmosis would generally induce an enhanced Th1 immune response with an increased production of inflammatory cytokines IFN-γ and IL-2 to limit infection. An analysis across T. gondii DNA vaccination studies reveals that the inducement of a Th1-type cellular immune response with significantly elevated levels of IFN-γ and IL-2 does not correlate with the degree of protective outcomes in mice. This means that the level of immunogenicity is not necessarily predictive of survival rates, as can be appreciated in several recent examples [33,34,35,36]. However, DNA vaccinations in general has shown to be effective at inducing the activation and proliferation of CD4+ and CD8+ T cells, along with eliciting specific antibodies essential for control of chronic infection.
The drawback of DNA vaccines is that the immune responses it generates are usually too weak to be significantly protective when administered in higher primates and humans [37]. Improvement in its potency can be gained by strategies such as codon optimization for increased protein expression, co-expression of cytokines, and molecular adjuvants, and using a heterologous prime-boost strategy [7,38]. In addition, DNA vaccination strategies using a combination of multiple antigens have been shown to be more effective in enhancing protective immunity than single subunit antigens [reviewed in 11]. A case in point is the antigenic combination of micronemal proteins (MIC2, MIC3, MIC4, M2AP, and AMA1) which achieved significant protection of 84% reduction in the brain tissue cyst burden and longer survival durations than single subunit antigen vaccination [39]. To ensure immunogenicity of the vaccines, the authors formulated a DNA vaccine based on known T-cell epitopes instead of using full-length antigens. This highlights that the identification of immunogenic epitopes capable of eliciting humoral and cellular immune responses is another important parameter to consider in vaccine design. The in silico identification of these functional epitopes seems increasingly possible in recent times via predictive binding algorithms for humans' MHC I molecules [8, reviewed in 40].
Co-administration of adjuvants has also shown to be a useful approach in boosting the immune response in DNA vaccinations to therapeutic levels. A recent example of this is the MIC8 vaccination studies. The MIC8 gene previously garnered a lot of interest due to the completely defective invasion phenotype of MIC8-parasite knockout mutant [41]. However, DNA vaccination with MIC8 alone generated only a modest increase in survival time [42]. In a strategy to enhance the protective immunity from MIC8 vaccination, co-administration of mIL-21 and mIL-15 cytokines showed improved survival time from 10.3±0.9 days (without cytokines) to 16.2±1.3 days (with cytokines) [43].
The highest protection achieved with DNA vaccination against toxoplasmosis was obtained by immunization with a plasmid encoding SAG, which conferred 100% survival in CH3 mice and 80% survival in BALB/c mice for at least 6 months post-lethal challenge [44]. The potency of this protective immunity exceeded a similar experiment using SAG1 protein vaccine which prolonged mouse survival, but couldn't prevent complete mortality [45]. However, several more recent studies using either the SAG1 antigen alone or in combination with other antigens showed less robust protective immunity, where the highest survival rate was 40%, even with the use of the same strain of parasite and mouse host [46,47,48]. This ambiguity may be attributed to the use of different SAG1 epitopes in each study. Another immunization study which also demonstrated a significant survival rate of 100% and 40% in BALB/c and C57BL/6 mice respectively following a lethal challenge, used a multi-epitope DNA vaccination [49]. This multi-epitope vaccine comprising 6 antigenic epitopes from SAG1, GRA1, GRA2 and GRA4 antigens also showed effective stimulation of a protective Th1 immune response; and forms part of an accumulating body of evidence on the efficacy of multi-antigen vaccinations against a complex parasite such T. gondii [49,50,51].
Over the past 4 years, toxoplasmosis DNA vaccination studies expanded to the use of many new parasite antigens, several of which yielded promising protective outcomes (Table 2). Among these latest developments, several single-antigen vaccines were successful in producing survivors from a lethal infection challenge, such as ROP8, TgCyP, and GRA6 with a range of survival rate between 37.5-50.0% [52,53,54]. Another subset of these DNA vaccine studies produced a significant increase in survival time (≥3 weeks) but no survivors following a lethal challenge, which was the ROP13, ROP16, ROP18, MIC13, and eIF4A [34,36,55,56,57]. There was a less significant survival outcome (≤2 weeks) from yet another subset of antigens, such as TgROM1, TgPLP1, TgCDPK3, and TgCDPK5 [33,58,59,60]. For effective comparisons, these recent studies in DNA vaccinations against toxoplasmosis are summarized in Table 2. Taken together, the next generation of vaccine design could include the empirical evaluation of functional antigenic epitope combinations in conjunction with immunological adjuvants for maximal efficacy.

LIVE, ATTENUATED VECTORS

Another vaccination strategy against toxoplasmosis involves the use of live, attenuated vectors, such as viruses and bacteria as vehicles to express recombinant T. gondii antigens in the host. The use of a live, attenuated vector to deliver T. gondii antigens provides the benefits of mimicry of the intracellular niche of the parasite by using innocuous recombinant virus or bacteria, and has been shown to be able to induce complete protection in some instances due to its intrinsic adjuvant properties. An example of this was shown in a DNA/viral vector heterologous prime-boost vaccination experiment. The GRA4 antigen delivered via recombinant attenuated vaccinia virus afforded 100% survival from infection with type II PLK/GFP tachyzoites, no detectable brain tissue cysts on 45 days post-infection, and significantly reduced brain parasite load [67]. In contrast, the plasmid-plasmid DNA vaccination group showed a lower survival rate of 70%. Some of the live vectors used in T. gondii vaccination studies include Salmonella typhimurium, pseudorabies virus, adenovirus, and modified vaccinia Ankara (MVA) (Table 3). The major advantages of live vectors as vaccine vehicles are its prospect as oral vaccines leading to inducement of mucosal immunity, as well as the potent immune response it elicits. These advantages has led to the development of these live vaccine formats for diseases ranging from cholera [68], to HIV infection [69] and cancer [70]. However, the use of recombinant, live vaccines faces the inevitable obstacles of safety risks and genetically-modified organisms (GMO) regulatory issues. The safety concerns that need to be addressed other than the more obvious ablation of virulence, is the lowering of risk of autoimmune responses and non-administration to immunocompromised individuals [reviewed in 71]. The vaccination studies using live, attenuated vectors against toxoplasmosis are summarized in Table 3.
Protective immunity to toxoplasmosis is highly complex as the interplay between the parasite's strain type and the host's genetic background influences disease pathogenesis [76,77]. The host's genetic influence over the selective processing of antigenic peptide, with the subsequent ability to present certain peptides determines disease severity in each individual. Therefore, the challenge of vaccine design is to determine protective epitopes across the antigenic diversity in T. gondii strains, capable of engaging with the diverse MHCs encompassing a majority of the human population to produce protective immunity [40]. Vaccine researchers now have an additional resource for testing vaccines by referring to the toxoplasmosis infection and pathogenesis model developed by Dubey et al. [78]. This in-depth study details infection responses across different mouse strains and various T. gondii strains, including the atypical strains. Thus, this model can be a highly useful tool in testing vaccine efficacy by providing a foundational standard for comparison.
In considering the platforms for vaccine delivery from the myriad of strategies ranging from DNA to live vaccines, it is worth noting that each platform presents its own benefits and disadvantages that are beyond the scope of this article to delve into. The interested reader is referred to an excellent review by Bruna-Romero et al. [79] on this topic. The development of pre-emptive vaccines remains a highly important pursuit in light of the persistent nature of chronic toxoplasmosis that is refractory to current drug treatments, and the increasing mortality associated with reactivated infections.

PASSIVE IMMUNIZATION

In the acute stage of toxoplasmosis, the host's innate immune response plays a key role in the control of infection [80], while long-term protective immunity is mediated by the adaptive immune response that drives it into a quiescent chronic stage but does not completely eradicate the parasite [81,82]. The persistent chronic stage can be lethal in reactivated infections affecting immunocompromised hosts. Thus, passive immunization could be a useful means of limiting clinical disease in hosts with reduced cellular immunity.
A major cell subset critical in disease control is the CD8+ T cells which produces the cytokine IFN-γ [9,10]. It was revealed that the adoptive transfer of immune CD8+ T cells to naïve animals could confer protective immunity and prolong survival in acute infections [82]. Although these are encouraging findings, translation of the application of syngeneic cellular immunotherapeutics to the masses could be challenging. Nonetheless, studies on passive cellular immunization could still provide invaluable insights on the factors affecting long-term immune control of toxoplasmosis. The inbred C57BL/6 mouse strain is mostly used in such studies due to its susceptibility to succumb to chronic toxoplasmosis, even with avirulent strains. This condition mimics reactivation of latent infections arising from immune compromise. One of the recent and interesting findings of this is that adoptive transfer of immune CD8+ T cells to pre-recrudescent chronically-infected C57BL/6 mice could provide transient protection from disease reactivation for up to 4 weeks post-treatment [83]. However, this same treatment could not rescue endogenous CD8+ T cells from functional exhaustion leading to apoptosis and the impairment of memory T cells development. Thus, long-term protective immunity was not achieved. These results seem to suggest that adoptive cellular immunotherapy provides significant protection in acute toxoplasmosis, but not so in a model of disease reactivation in chronic toxoplasmosis.
While CD8+ T cells and IFN-γ are important mediators of the host immune responses to resist T. gondii infection, antibodies produced by B cells are essential for long-term resistance to the disease by the production of specific antibodies [84,85]. The absence of antibody production in the host exacerbates infection and results in chronic stage mortality. In a study using CD4+ depleted mice to parallel CD4+ deficiency in AIDS patients and other immunocompromised individuals, passive immunization with immune serum has shown to be able to prolong survival and transiently overcome impaired disease resistance [86]. This suggests that the production of specific antibodies together with cellular immune responses is an important feature in the generation of adaptive immunity to T. gondii. Therefore, the potential development of antibodies as immunotherapeutic agents to arrest disease progression would prove highly beneficial to individuals suffering from impaired immunity.
In an experiment using Fab antibody fragments specific to T. gondii SAG1 antigen, it was shown that the antibodies blocked parasites attachment to host cells in vitro by 52% at an antibody concentration of 200 µg/ml [87]. Passive immunization of mice with 10 mg of this antibody provided improved survival rates of up to 50% following lethal challenge infection [87]. This survival rate improvement is a significant finding considering that the Fab fragments used in this study is lacking the Fc region of full antibodies which mediates Fc-receptor-dependent phagocytosis of opsonized tachyzoites, cellular cytotoxicity and complement activation. Therefore, it would seem that the antibodies mediate its protective function by binding to the parasites' surface and directly blocking infection of host cells, which is consistent with an observation by Sayles and co-workers in their study [85].
Passive immunization with anti-GRA2 and anti-GRA6 monoclonal antibodies have also resulted in inhibition of parasite invasion and enhanced survival times in mice. However, the anti-SAG1 antibody yielded a more significant protective immunity than the antibodies targeted to the dense granules antigens [88]. Another passive immunization experiment using monoclonal antibodies against nucleoside triphosphate hydrolase-II (NTPase-II) showed no inhibition of parasite invasion, but a significant reduction in T. gondii replication and promoted prolonged survival in mice after a lethal challenge [89]. Taken together, the development of anti-toxoplasmosis antibodies providing greater protection from host infection could benefit from a combinatorial antibody formulation that blocks parasite attachment, invasion and proliferation; and thus warrants further investigations.
Recently, polyclonal antibodies against T. gondii was produced in egg yolks from immunized chicken and showed a comparable avidity and reactivity profile to mammalian-derived antibodies [90]. The main advantage of using a chicken platform is its' very high yield of antibodies (4 mg/ml purified IgY) which can be purified inexpensively and obviates the need to bleed animals. Because of the phylogenetic distance between avian and mammalian species, chicken IgY antibodies has the inherent biochemical advantage of not activating the mammalian complement system, and does not react with rheumatoid factors and anti-human IgG antibodies [91]. However, the therapeutic effect of these avian antibodies remains uncertain and requires further study. In conclusion, regardless of the production platform for anti-toxoplasmosis antibodies, therapeutic antibodies to ameliorate the significant disease morbidity and mortality associated with reactivated infections is obviously needed and evidence from past studies shows that this can be a feasible toxoplasmosis treatment for immunocompromised patients.

CONCLUDING REMARKS

The immune evasive nature and complex life cycle of T. gondii has made the design of immunotherapeutics aimed at providing complete protection a daunting challenge. Generally, vaccine studies thus far have shown that multi-antigenic formulations confer better protection than single-subunit vaccines. At this juncture, research on adjuvants for vaccine co-administration is also crucial as alum has been the only one out of five licensed human adjuvants tested with toxoplasmosis vaccine formulations. In context of toxoplasmosis vaccination, this isn't an ideal option as alum augments the Th2 immune response, instead of the predominantly Th1 responses usually seen in protective immunity. Therefore, studies on multiple combinations of antigens with co-delivery of adjuvants or cytokines may yield better protective outcomes and progressively narrow the gap towards achieving sterile immunity. Vaccines and immunotherapeutic antibodies targeted to the parasite's surface membrane antigens present highly promising prospects considering the high immunogenicity of these antigens and the resulting significant parasitaemia inhibition from multiple studies reviewed thus far. Therefore, immunotherapeutics targeting either a single or multiple parasite surface antigens, in combination with invasion-related antigens such as the MIC, AMA1 and ROP proteins can be a potential strategy for generating a neutralizing immune response against toxoplasmosis. The discovery of atypical strains in the South American region which are more virulent than the classic European types I, II and III strains, and causes severe illness; would mean that next generation research efforts should also consider designing assays for cross-protection evaluation. These research efforts could be aided by a presently available model of toxoplasmosis infection strains in mouse [78]. A universal or multi-type anti-T. gondii immunotherapeutics is a highly valuable goal for global disease control. To this end, the completion of the genomes of predominant strains of T. gondii together with bioinformatics algorithms to predict MHC I-binding peptides could be a valuable tool to inform the design of broadly protective immunotherapeutics.
University of MalayaUM.C/625/1/HIR/MOHE/SCI/18

ACKNOWLEDGMENTS

Work related to the preparation of this review was supported by University of Malaya, Malaysia (grant: UM.C/625/1/HIR/MOHE/SCI/18).

Conflict of interest

We have no conflict of interest related to this review article.

REFERENCES

1. Chowdhury M. Toxoplasmosis: a review. J Med 1986;17:373-396. PMID: 3295094.

2. Brindle R, Holliman R, Gilks C, Waiyaki P. Toxoplasma antibodies in HIV-positive patients from Nairobi. Trans R Soc Trop Med Hyg 1991;85:750-751. PMID: 1801345.
crossref
3. Mui EJ, Schiehser GA, Milhous WK, Hsu H, Roberts CW, Kirisits M, Muench S, Rice D, Dubey JP, Fowble JW, Rathod PK, Queener SF, Liu SR, Jacobus DP, McLeod R. Novel triazine JPC-2067-B inhibits Toxoplasma gondii in vitro and in vivo. PLoS Negl Trop Dis 2008;2:e190. PMID: 18320016.
crossref pmid pmc
4. Khan A, Jordan C, Muccioli C, Vallochi AL, Rizzo LV, Belfort R Jr, Vitor RWA, Silveira C, Sibley LD. Genetic Divergence of Toxoplasma gondii Strains Associated with Ocular Toxoplasmosis, Brazil. Emerg Infect Dis 2006;12:942-949. PMID: 16707050.
crossref pmid pmc
5. Bottos J, Miller RH, Belfort RN, Macedo AC; UNIFESP Toxoplasmosis Group, Belfort R Jr, Grigg ME. Bilateral retinochoroiditis caused by an atypical strain of Toxoplasma gondii. Br J Ophthalmol 2009;93:1546-1550. PMID: 19666926.
crossref pmid pmc
6. Garcia J, Innes EA, Katzer F. Current progress toward vaccines against Toxoplasma gondii. Vaccine Devel Ther 2014;4:23-37.
crossref
7. Kur J, Holec-Gasior L, Hiszczynska-Sawicka E. Current status of toxoplasmosis vaccine development. Expert Rev Vaccines 2009;8:791-808. PMID: 19485758.
crossref pmid
8. Tan TG, Mui E, Cong H, Witola W, Montpetit A, Muench SP, Sidney J, Alexander J, Sette A, Grigg M, Maewal A, McLeod R. Identification of T. gondii epitopes, adjuvants, and host genetic factors that influence protection of mice and humans. Vaccine 2010;28:3977-3989. PMID: 20347630.
crossref pmid pmc
9. Gazzinelli RT, Hakim FT, Hieny S, Shearer GM, Sher A. Synergistic role of CD4+ and CD8+ T lymphocytes in IFN-gamma production and protective immunity induced by an attenuated Toxoplasma gondii vaccine. J Immunol 1991;146:286-292. PMID: 1670604.
pmid
10. Brown CR, McLeod R. Class I MHC genes and CD8+ T cells determine cyst number in Toxoplasma gondii infection. J Immunol 1990;145:3438-3441. PMID: 2121825.
pmid
11. Jongert E, Roberts CW, Gargano N, Förster-Waldl E, Petersen E. Vaccines against Toxoplasma gondii: challenges and opportunities. Mem Inst Oswaldo Cruz 2009;104:252-266. PMID: 19430651.
crossref pmid
12. Debard N, Buzoni-Gatel D, Bout D. Intranasal immunization with SAG1 protein of Toxoplasma gondii in association with cholera toxin dramatically reduces development of cerebral cysts after oral infection. Infect Immun 1996;64:2158-2166. PMID: 8675321.
pmid pmc
13. Khan IA, Ely KH, Kasper LH. A purified parasite antigen (p30) mediates CD8+ T cell immunity against fatal Toxoplasma gondii infection in mice. J Immunol 1991;147:3501-3506. PMID: 1940350.
pmid
14. Bonenfant C, Dimier-Poisson I, Velge-Roussel F, Buzoni-Gatel D, Del Giudice G, Rappuoli R, Bout D. Intranasal immunization with SAG1 and nontoxic mutant heat-labile enterotoxins protects mice against Toxoplasma gondii. Infect Immun 2001;69:1605-1612. PMID: 11179334.
crossref pmid pmc
15. Qu D, Han J, Du A. Enhancement of protective immune response to recombinant Toxoplasma gondii ROP18 antigen by ginsenoside Re. Exp Parasitol 2013;135:234-239. PMID: 23896123.
crossref pmid
16. Letscher-Bru V, Pfaff AW, Abou-Bacar A, Filisetti D, Antoni E, Villard O, Klein JP, Candolfi E. Vaccination with Toxoplasma gondii SAG-1 protein is protective against congenital toxoplasmosis in BALB/c mice but not in CBA/J mice. Infect Immun 2003;71:6615-6619. PMID: 14573684.
crossref pmid pmc
17. Chuang SC, Ko JC, Chen CP, Du JT, Yang CD. Induction of long-lasting protective immunity against Toxoplasma gondii in BALB/c mice by recombinant surface antigen 1 protein encapsulated in poly (lactide-co-glycolide) microparticles. Parasit Vectors 2013;6:34. PMID: 23398973.
crossref
18. Chuang SC, Ko JC, Chen CP, Du JT, Yang CD. Encapsulation of chimeric protein rSAG1/2 into poly (lactide-co-glycolide) microparticles induces long-term protective immunity against Toxoplasma gondii in mice. Exp Parasitol 2013;134:430-437. PMID: 23624036.
crossref pmid
19. Yin LT, Hao HX, Wang HL, Zhang JH, Meng XL, Yin GR. Intranasal immunisation with recombinant Toxoplasma gondii actin partly protects mice against Toxoplasmosis. PLoS ONE 2013;8:e82765. PMID: 24386114.
crossref pmid pmc
20. Wang HL, Pang M, Yin LT, Zhang JH, Meng XL, Yu BF, Guo R, Bai JZ, Zheng GP, Yin GR. Intranasal immunisation of the recombinant Toxoplasma gondii receptor for activated C kinase 1 partly protects mice against T. gondii infection. Acta Trop 2014;137:58-66. PMID: 24813415.
crossref pmid
21. Nakaar V, Ngô HM, Aaronson EP, Coppens I, Stedman TT, Joiner KA. Pleiotropic effect due to targeted depletion of secretory rhoptry protein ROP2 in Toxoplasma gondii. J Cell Sci 2003;116:2311-2320. PMID: 12711703.
crossref pmid
22. Nam HW. GRA Proteins of Toxoplasma gondii: maintenance of host-parasite interactions across the parasitophorous vacuolar membrane. Korean J Parasitol 2009;47(suppl):S29-S37. PMID: 19885333.
crossref pmid pmc
23. Martin V, Supanitsky A, Echeverria PC, Litwin S, Tanos T, Roodt ARD, Guarnera EA, Angel SO. Recombinant GRA4 or ROP2 protein combined with alum or the gra4 gene provides partial protection in chronic murine models of toxoplasmosis. Clin Diagn Lab Immunol 2004;11:704-710. PMID: 15242945.
crossref
24. Dziadek B, Gatkowska J, Brzostek A, Dziadek J, Dzitko K, Grzybowski M, Dlugonska H. Evaluation of three recombinant multi-antigenic vaccines composed of surface and secretory antigens of Toxoplasma gondii in murine models of experimental toxoplasmosis. Vaccine 2011;29:821-830. PMID: 21087690.
crossref pmid
25. Dziadek B, Gatkowska J, Grzybowski M, Dziadek J, Dzitko K, Dlugonska H. Toxoplasma gondii: the vaccine potential of three trivalent antigen-cocktails composed of recombinant ROP2, ROP4, GRA4 and SAG1 proteins against chronic toxoplasmosis in BALB/c mice. Exp Parasitol 2012;131:133-138. PMID: 22445587.
crossref pmid
26. El Bissati K, Zhou Y, Dasgupta D, Cobb D, Dubey JP, Burkhard P, Lanar DE, McLeod R. Effectiveness of a novel immunogenic nanoparticle platform for Toxoplasma peptide vaccine in HLA transgenic mice. Vaccine 2014;32:3243-3248. PMID: 24736000.
crossref pmid pmc
27. Del L. Yácono M, Farran I, Becher ML, Sander V, Sánchez VR, Martín V, Veramendi J, Clemente M. A chloroplast-derived Toxoplasma gondii GRA4 antigen used as an oral vaccine protects against toxoplasmosis in mice. Plant Biotechnol J 2012;10:1136-1144. PMID: 23020088.
crossref pmid
28. Laguia-Becher M, Martin V, Kraemer M, Corigliano M, Yacono ML, Goldman A, Clemente M. Effect of codon optimization and subcellular targeting on Toxoplasma gondii antigen SAG1 expression in tobacco leaves to use in subcutaneous and oral immunization in mice. BMC Biotechnol 2010;10:52. PMID: 20633272.
crossref pmid pmc
29. Zheng B, Lu S, Tong Q, Kong Q, Lou D. The virulence-related rhoptry protein 5 (ROP5) of Toxoplasmag gondii is a novel vaccine candidate against toxoplasmosis in mice. Vaccine 2013;31:4578-4584. PMID: 23928460.
crossref pmid
30. Sanchez VR, Pitkowski MN, Fernandez Cuppari AV, Rodriguez FM, Fenoy IM, Frank FM, Goldman A, Corral RS, Martin V. Combination of CpG-oligodeoxynucleotides with recombinant ROP2 or GRA4 proteins induces protective immunity against Toxoplasma gondii infection. Exp Parasitol 2011;128:448-453. PMID: 21554876.
crossref pmid
31. Wang Y, Wang M, Wang G, Pang A, Fu B, Yin H, Zhang D. Increased survival time in mice vaccinated with a branched lysine multiple antigenic peptide containing B- and T-cell epitopes from T. gondii antigens. Vaccine 2011;29:8619-8623. PMID: 21939715.
crossref pmid
32. Wang HL, Li YQ, Yin LT, Meng XL, Guo M, Zhang JH, Liu HL, Liu JJ, Yin GR. Toxoplasma gondii Protein Disulfide Isomerase (TgPDI) is a novel vaccine candidate against toxoplasmosis. PLoS ONE 2013;8:e70884. PMID: 23967128.
crossref pmid pmc
33. Zhang NZ, Huang SY, Xu Y, Chen J, Wang JL, Tian WP, Zhu XQ. Evaluation of immune responses in mice after DNA immunization with putative Toxoplasma gondii Calcium-Dependent Protein Kinase 5 (TgCDPK5). Clin Vaccine Immunol 2014;21:924-929. PMID: 24789795.
crossref pmid pmc
34. Yuan ZG, Ren D, Zhou DH, Zhang XX, Petersen E, Li XZ, Zhou Y, Yang GL, Zhu XQ. Evaluation of protective effect of pVAX-TgMIC13 plasmid against acute and chronic Toxoplasma gondii infection in a murine model. Vaccine 2013;31:3135-3139. PMID: 23707448.
crossref pmid
35. Chen J, Zhou DH, Li ZY, Petersen E, Huang SY, Song HQ, Zhu XQ. Toxoplasma gondii: protective immunity induced by rhoptry protein 9 (TgROP9) against acute toxoplasmosis. Exp Parasitol 2014;139:42-48. PMID: 24602875.
crossref pmid
36. Chen J, Huang SY, Li ZY, Yuan ZG, Zhou DH, Petersen E, Zhang NZ, Zhu XQ. Protective immunity induced by a DNA vaccine expressing eIF4A of Toxoplasma gondii against acute toxoplasmosis in mice. Vaccine 2013;31:1734-1739. PMID: 23370151.
crossref pmid
37. Liu Q, Singla LD, Zhou H. Vaccines against Toxoplasma gondii: status, challenges and future directions. Hum Vaccin Immunother 2012;8:1305-1308. PMID: 22906945.
crossref pmid pmc
38. Saade F, Petrovsky N. Technologies for enhanced efficacy of DNA vaccines. Expert Rev Vaccines 2012;11:189-209. PMID: 22309668.
crossref pmid pmc
39. Beghetto E, Nielsen HV, Porto PD, Buffolano W, Guglietta S, Felici F, Petersen E, Gargano N. A combination of antigenic regions of Toxoplasma gondii microneme proteins induces protective immunity against oral infection with parasite cysts. J Infect Dis 2005;191:637-645. PMID: 15655789.
crossref pmid
40. Henriquez FL, Woods S, Cong H, McLeod R, Roberts CW. Immunogenetics of Toxoplasma gondii informs vaccine design. Trends Parasitol 2010;26:550-555. PMID: 20580611.
crossref pmid
41. Kessler H, Herm-Gotz A, Hegge S, Rauch M, Soldati-Favre D, Frischknecht F, Meissner M. Microneme protein 8-a new essential invasion factor in Toxoplasma gondii. J Cell Sci 2008;121:947-956. PMID: 18319299.
crossref pmid
42. Liu MM, Yuan ZG, Peng GH, Zhou DH, He XH, Yan C, Yin CC, He Y, Lin RQ, Song HQ, Zhu XQ. Toxoplasma gondii microneme protein 8 (MIC8) is a potential vaccine candidate against toxoplasmosis. Parasitol Res 2010;106:1079-1084. PMID: 20177910.
crossref pmid
43. Li ZY, Chen J, Petersen E, Zhou DH, Huang SY, Song HQ, Zhu XQ. Synergy of mIL-21 and mIL-15 in enhancing DNA vaccine efficacy against acute and chronic Toxoplasma gondii infection in mice. Vaccine 2014;32:3058-3065. PMID: 24690150.
crossref pmid
44. Nielsen HV, Lauemøller SL, Christiansen L, Buus S, Fomsgaard A, Petersen E. Complete protection against lethal Toxoplasma gondii infection in mice immunized with a plasmid encoding the SAG1 gene. Infect Immun 1999;67:6358-6363. PMID: 10569750.
pmid pmc
45. Petersen E, Nielsen HV, Christiansen L, Spenter J. Immunization with E. coli produced recombinant T. gondii SAG1 with alum as adjuvant protect mice against lethal infection with Toxoplasma gondii. Vaccine 1998;16:1283-1289. PMID: 9682392.
crossref pmid
46. Cong H, Zhang M, Xin Q, Wang Z, Li Y, Zhao Q, Zhou H, He S. Compound DNA vaccine encoding SAG1/SAG3 with A2/B subunit of cholera toxin as a genetic adjuvant protects BALB/c mice against Toxoplasma gondii. Parasit Vectors 2013;6:63. PMID: 23497561.
crossref pmid pmc
47. Meng M, He S, Zhao G, Bai Y, Zhou H, Cong H, Lu G, Zhao Q, Zhu XQ. Evaluation of protective immune responses induced by DNA vaccines encoding Toxoplasma gondii surface antigen 1 (SAG1) and 14-3-3 protein in BALB/c mice. Parasit Vectors 2012;5:273. PMID: 23181694.
crossref pmid pmc
48. Zhou H, Min J, Zhao Q, Gu Q, Cong H, Li Y, He S. Protective immune response against Toxoplasma gondii elicited by a recombinant DNA vaccine with a novel genetic adjuvant. Vaccine 2012;30:1800-1806. PMID: 22240340.
crossref pmid
49. Liu S, Shi L, Cheng YB, Fan GX, Ren HX, Yuan YK. Evaluation of protective effect of multi-epitope DNA vaccine encoding six antigen segments of Toxoplasma gondii in mice. Parasitol Res 2009;105:267-274. PMID: 19288132.
crossref pmid
50. Fachado A, Rodriguez A, Angel SO, Pinto DC, Vila I, Acosta A, Amendoeira RR, Lannes-Vieira J. Protective effect of a naked DNA vaccine cocktail against lethal toxoplasmosis in mice. Vaccine 2003;21:1327-1335. PMID: 12615427.
crossref pmid
51. Ivory C, Chadee K. DNA vaccines: designing strategies against parasitic infections. Genet Vaccines Ther 2004;2:17. PMID: 15579202.
crossref pmid pmc
52. Parthasarathy S, Fong MY, Ramaswamy K, Lau YL. Protective immune response in BALB/c mice induced by DNA vaccine of the ROP8 gene of Toxoplasma gondii. Am J Trop Med Hyg 2013;88:883-887. PMID: 23509124.
crossref pmid pmc
53. Gong P, Huang X, Yu Q, Li Y, Huang J, Li J, Yang J, Li H, Zhang G, Ren W, Zhang X. The protective effect of a DNA vaccine encoding the Toxoplasma gondii cyclophilin gene in BALB/c mice. Parasite Immunol 2013;35:140-146. PMID: 23278173.
crossref
54. Sun XM, Zou J, AA ES, Yan WC, Liu XY, Suo X, Wang H, Chen QJ. DNA vaccination with a gene encoding Toxoplasma gondii GRA6 induces partial protection against toxoplasmosis in BALB/c mice. Parasit Vectors 2011;4:213. PMID: 22070984.
crossref pmid pmc
55. Wang PY, Yuan ZG, Petersen E, Li J, Zhang XX, Li XZ, Li HX, Lv ZC, Cheng T, Ren D, Yang GL, Lin RQ, Zhu XQ. Protective efficacy of a Toxoplasma gondii Rhoptry Protein 13 plasmid DNA vaccine in mice. Clin Vaccine Immunol 2012;19:1916-1920. PMID: 23015648.
crossref pmid pmc
56. Yuan ZG, Zhang XX, He XH, Petersen E, Zhou DH, He Y, Lin RQ, Li XZ, Chen XL, Shi XR, Zhong XL, Zhang B, Zhu XQ. Protective immunity induced by Toxoplasma gondii rhoptry protein 16 against toxoplasmosis in mice. Clin Vaccine Immunol 2011;18:119-124. PMID: 21106780.
crossref pmid pmc
57. Yuan ZG, Zhang XX, Lin RQ, Petersen E, He S, Yu M, He XH, Zhou DH, He Y, Li HX, Liao M, Zhu XQ. Protective effect against toxoplasmosis in mice induced by DNA immunization with gene encoding Toxoplasma gondii ROP18. Vaccine 2011;29:6614-6619. PMID: 21762755.
crossref pmid
58. Li J, Han Q, Gong P, Yang T, Ren B, Li S, Zhang X. Toxoplasma gondii rhomboid protein 1 (TgROM1) is a potential vaccine candidate against toxoplasmosis. Vet Parasitol 2012;184:154-160. PMID: 21906881.
crossref pmid
59. Yan HK, Yuan ZG, Petersen E, Zhang XX, Zhou DH, Liu Q, He Y, Lin RQ, Xu MJ, Chen XL, Zhong XL, Zhu XQ. Toxoplasma gondii: protective immunity against experimental toxoplasmosis induced by a DNA vaccine encoding the perforin-like protein 1. Exp Parasitol 2011;128:38-43. PMID: 21310148.
crossref pmid
60. Zhang NZ, Huang SY, Zhou DH, Chen J, Xu Y, Tian WP, Lu J, Zhu XQ. Protective immunity against Toxoplasma gondii induced by DNA immunization with the gene encoding a novel vaccine candidate: calcium-dependent protein kinase 3. BMC Infect Dis 2013;13:512. PMID: 24176018.
crossref pmid pmc
61. Zhang M, Zhao L, Song J, Li Y, Zhao Q, He S, Cong H. DNA vaccine encoding the Toxoplasma gondii bradyzoite-specific surface antigens SAG2CDX protect BALB/c mice against type II parasite infection. Vaccine 2013;31:4536-4540. PMID: 23933373.
crossref pmid
62. Yu L, Yamagishi J, Zhang S, Jin C, Aboge GO, Zhang H, Zhang G, Tanaka T, Fujisaki K, Nishikawa Y, Xuan X. Protective effect of a prime-boost strategy with plasmid DNA followed by recombinant adenovirus expressing TgAMA1 as vaccines against Toxoplasma gondii infection in mice. Parasitol Int 2012;61:481-486. PMID: 22537971.
crossref pmid
63. Meng M, Zhou A, Lu G, Wang L, Zhao G, Han Y, Zhou H, Cong H, Zhao Q, Zhu XQ, He S. DNA prime and peptide boost immunization protocol encoding the Toxoplasma gondii GRA4 induces strong protective immunity in BALB/c mice. BMC Infect Dis 2013;13:494. PMID: 24148219.
crossref pmid pmc
64. Zhao H, Huang FY, Guo JL, Tan GH. Evaluation on the immune response induced by DNA vaccine encoding MIC8 co-immunized with IL-12 genetic adjuvant against Toxoplasma gondii infection. Zhongguo Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi 2013;31:284-289. PMID: 24812879.
pmid
65. Chen J, Huang SY, Zhou DH, Li ZY, Petersen E, Song HQ, Zhu XQ. DNA immunization with eukaryotic initiation factor-2α of Toxoplasma gondii induces protective immunity against acute and chronic toxoplasmosis in mice. Vaccine 2013;31:6225-6231. PMID: 24183979.
crossref pmid
66. Cui X, Lei T, Yang D, Hao P, Li B, Liu Q. Toxoplasma gondii immune mapped protein-1 (TgIMP1) is a novel vaccine candidate against toxoplasmosis. Vaccine 2012;30:2282-2287. PMID: 22310204.
crossref pmid
67. Zhang G, Huong VT, Battur B, Zhou J, Zhang H, Liao M, Kawase O, Lee EG, Dautu G, Igarashi M, Nishikawa Y, Xuan X. A heterologous prime-boost vaccination regime using DNA and a vaccinia virus, both expressing GRA4, induced protective immunity against Toxoplasma gondii infection in mice. Parasitology 2007;134:1339-1346. PMID: 17506929.
crossref
68. Taylor DN, Tacket CO, Losonsky G, Castro O, Gutierrez J, Meza R, Nataro JP, Kaper JB, Wasserman SS, Edelman R, Levine MM, Cryz SJ. Evaluation of a bivalent (CVD 103-HgR/CVD 111) live oral cholera vaccine in adult volunteers from the United States and Peru. Infect Immun 1997;65:3852-3856. PMID: 9284163.
pmid pmc
69. Virnik K, Hockenbury M, Ni Y, Beren J, Pavlakis GN, Felber BK, Berkower I. Live attenuated rubella vectors expressing SIV and HIV vaccine antigens replicate and elicit durable immune responses in rhesus macaques. Retrovirology 2013;10:99. PMID: 24041113.
crossref pmid pmc
70. Toussaint B, Chauchet X, Wang Y, Polack B, Le Gouellec A. Live-attenuated bacteria as a cancer vaccine vector. Expert Rev Vaccines 2013;12:1139-1154. PMID: 24124876.
crossref pmid
71. Detmer A, Glenting J. Live bacterial vaccines-a review and identification of potential hazards. Microb Cell Fact 2006;5:23. PMID: 16796731.
crossref pmid pmc
72. Mendes ÉA, Fonseca FG, Casério BM, Colina JP, Gazzinelli RT, Caetano BC. Recombinant vaccines against T. gondii: comparison between homologous and heterologous vaccination protocols using two viral vectors expressing SAG1. PLoS One 2013;8:e63201. PMID: 23690999.

73. Cong H, Gu QM, Jiang Y, He SY, Zhou HY, Yang TT, Li Y, Zhao QL. Oral immunization with a live recombinant attenuated Salmonella typhimurium protects mice against Toxoplasma gondii. Parasite Immunol 2005;27:29-35. PMID: 15813720.
crossref
74. Liu Q, Gao S, Jiang L, Shang L, Men J, Wang Z, Zhai Y, Xia Z, Hu R, Zhang X, Zhu XQ. A recombinant pseudorabies virus expressing TgSAG1 protects against challenge with the virulent Toxoplasma gondii RH strain and pseudorabies in BALB/c mice. Microbes Infect 2008;10:1355-1362. PMID: 18761418.
crossref pmid
75. Nie H, Fang R, Xiong BQ, Wang LX, Hu M, Zhou YQ, Zhao JL. Immunogenicity and protective efficacy of two recombinant pseudorabies viruses expressing Toxoplasma gondii SAG1 and MIC3 proteins. Vet Parasitol 2011;181:215-221. PMID: 21632181.
crossref pmid
76. Mack DG, Johnson JJ, Roberts F, Roberts CW, Estes RG, David C, Grumet CF, McLeod R. HLA-class II genes modify outcome of Toxoplasma gondii infection. Int J Parasitol 1999;29:1351-1358. PMID: 10579423.
crossref pmid
77. Suzuki Y, Wong SY, Grumet FC, Fessel J, Montoya JG, Zolopa AR, Portmore A, Schumacher-Perdreau F, Schrappe M, Koppen S, Ruf B, Brown BW, Remington JS. Evidence for genetic regulation of susceptibility to toxoplasmic encephalitis in AIDS patients. J Infect Dis 1996;173:265-268. PMID: 8537674.
crossref pmid
78. Dubey JP, Ferreira LR, Martins J, McLeod R. Oral oocyst-induced mouse model of toxoplasmosis: effect of infection with Toxoplasma gondii strains of different genotypes, dose, and mouse strains (transgenic, out-bred, in-bred) on pathogenesis and mortality. Parasitology 2012;139:1-13. PMID: 22078010.
crossref pmid pmc
79. Bruna-Romero O, de Oliveira DM, de Andrade-Neto VF. Toxoplasmosis: Advances and Vaccine Perspectives. In Rodriguez-Morales AJ ed, Current Topics in Tropical Medicine. Rijeka. Croa tia. InTech. 2012.

80. Yarovinsky F. Innate immunity to Toxoplasma gondii infection. Nat Rev Immunol 2014;14:109-121. PMID: 24457485.
crossref pmid
81. Beghetto E, Spadoni A, Buffolano W, Del Pezzo M, Minenkova O, Pavoni E, Pucci A, Cortese R, Felici F, Gargano N. Molecular dissection of the human B-cell response against Toxoplasma gondii infection by lambda display of cDNA libraries. Int J Parasitol 2003;33:163-173. PMID: 12633654.
crossref pmid
82. Bhadra R, Gigley JP, Khan IA. The CD8 T-cell road to immunotherapy of toxoplasmosis. Immunotherapy 2011;3:789-801. PMID: 21668315.
crossref pmid pmc
83. Bhadra R, Cobb DA, Khan IA. Donor CD8+ T cells prevent Toxoplasma gondii de-encystation but fail to rescue the exhausted endogenous CD8+ T cell population. Infect Immun 2013;81:3414-3425. PMID: 23817617.
crossref pmid pmc
84. Kang H, Remington JS, Suzuki Y. Decreased resistance of B cell-deficient mice to infection with Toxoplasma gondii despite unimpaired expression of IFN-γ, TNF-α, and inducible nitric oxide synthase. J Immunol 2000;164:2629-2634. PMID: 10679102.
crossref pmid
85. Sayles PC, Gibson GW, Johnson LL. B cells are essential for vaccination-induced resistance to virulent Toxoplasma gondii. Infect Immun 2000;68:1026-1033. PMID: 10678903.
crossref pmid pmc
86. Johnson LL, Sayles PC. Deficient humoral responses underlie susceptibility to Toxoplasma gondii in CD4-deficient mice. Infect Immun 2002;70:185-191. PMID: 11748181.
crossref pmid pmc
87. Fu YF, Feng M, Ohnishi K, Kimura T, Itoh J, Cheng XJ, Tachibana H. Generation of a neutralizing human monoclonal antibody Fab fragment to surface antigen 1 of Toxoplasma gondii tachyzoites. Infect Immun 2011;79:512-517. PMID: 20956568.
crossref pmid pmc
88. Cha DY, Song IK, Lee GS, Hwang OS, Noh HJ, Yeo SD, Shin DW, Lee YH. Effects of specific monoclonal antibodies to dense granular proteins on the invasion of Toxoplasma gondii in vitro and in vivo. Korean J Parasitol 2001;39:233-240. PMID: 11590913.
crossref pmid pmc
89. Tan F, Hu X, Pan CW, Ding JQ, Chen XG. Monoclonal antibodies against nucleoside triphosphate hydrolase-II can reduce the replication of Toxoplasma gondii. Parasitol Int 2010;59:141-146. PMID: 20056166.
crossref pmid
90. Ferreira à Jr, Santiago FM, Silva MV, Ferreira FB, Macedo AG Jr, Mota CM, Faria MS, Silva Filho HH, Silva DA, Cunha-Júnior JP, Mineo JR, Mineo TW. Production, characterization and applications for Toxoplasma gondii-specific polyclonal chicken egg yolk immunoglobulins. PLoS ONE 2012;7:e40391. PMID: 22808150.
crossref pmid pmc
91. Larsson A, Balow RM, Lindahl TL, Forsberg PO. Chicken antibodies: taking advantage of evolution-a review. Poult Sci 1993;72:1807-1812. PMID: 8415358.
crossref pmid
Table 1.
Examples of immunization with protein vaccines against Toxoplasma gondii
Antigen Antigen delivery Mouse strain Challenge Results Study
ROP5 Protein (100 µg)+FCA+2 boosters in IFA, sc BALB/c 100 tachyzoites, RH strain Increased survival time relative to controls, but no complete protection, complete mortality by day 12 [29]
ROP5+SAG1 Proteins (100 µg each)+FCA+2 boosters in IFA, sc BALB/c 100 tachyzoites, RH strain Increased survival time (12.1 ± 3.4 days). No complete protection, complete mortality by day 17. Mixed Th1/Th2 immune response. [29]
ROP2+GRA4+SAG1 Proteins (total amount: 10 µg)+IFA, sc C3H/HeJ (H-2k) and C57BL/6 (H-2b) 5 tissue cysts, DX (Type II) strain C3H/HeJ: No significant reduction in brain tissue cysts [24]
C57BL/6: Reduced brain tissue cysts (55%)
ROP2+ROP4+SAG1 Proteins (total amount: 10 µg)+IFA, sc C3H/HeJ (H-2k) and C57BL/6 (H-2b) 5 tissue cysts, DX (Type II) strain C3H/HeJ: Reduced brain tissue cysts (71%) [24]
C57BL/6: Reduced brain tissue cysts (90%)
ROP2+ROP4+GRA4 Proteins (total amount: 10 µg)+IFA, sc C3H/HeJ (H-2k) and C57BL/6 (H-2b) 5 tissue cysts, DX (Type II) strain C3H/HeJ: Reduced brain tissue cysts (59%) [24]
C57BL/6: Reduced brain tissue cysts (41%)
Reduced parasitaemia to 71% (C3H/HeJ) and 90% (C57BL/6)
ROP2+ROP4+SAG1 Proteins (total amount: 10 µg)+IFA, sc BALB/c (H-2d) 5 tissue cysts, DX (Type II) strain Reduced brain tissue cysts (77%) [25]
ROP2+ROP4+GRA4 Proteins (total amount: 10 µg)+IFA, sc BALB/c (H-2d) 5 tissue cysts, DX (Type II) strain Reduced brain tissue cysts (84%) [25]
ROP2 Protein (10 µg)+CpG (10 µg), im C3H/HeN (H-2k) 20 tissue cysts, Me49 (Type II) strain Reduced brain tissue cysts (63%) [30]
ROP2+GRA4 Proteins (10 µg each)+CpG (10 µg), im C3H/HeN (H-2k) 20 tissue cysts, Me49 (Type II) strain Reduced brain tissue cysts (66%) [30]
SAG1 Protein (10 µg)+PLG encapsulationa, ip BALB/c 1 × 104 tachyzoites, RH strain Increased survival (endpoint: 28 days) of 80% [17]
Control immunized group survival was 20%
SAG1+SAG2 Protein (10 µg)+PLG encapsulation, ip BALB/c 1 × 104 tachyzoites, RH strain Increased survival (endpoint: 28 days) of 83% [18]
SAG1+GRA1+GRA4 (T- and B-cell epitopes) Protein (50 µg)+FCA+2 boosters in IFA, im BALB/c and Kunming 10 tachyzoites, GJS (Type I) strain BALB/c: Increased survival time (18.3 ± 0.9 days) [31]
Kunming: Increased survival time (16.0 ± 0.8 days)
GRA4 Protein from leaf extracts of GRA4- recombinant plants (0.5 µg)b, oral C57BL/6 20 tissue cysts, Me49 (Type II) strain Reduced brain tissue cysts relative to control (59%) [27]
Increased Th1/Th2 cytokine
TgACTc Protein (30 µg), in BALB/c Acute infection: 4 × 104 tachyzoites, RH strain Increased survival rate of 50% relative to control [19]
Chronic infection: 1 × 104 tachyzoites, RH strain Reduced tachyzoites load in liver (60.05%) and brain (49.75%)
TgPDId Protein (30 µg), in BALB/c Acute infection: 4 × 104 tachyzoites, RH strain Increased survival rate of 31% relative to control [32]
Chronic infection: 1 × 104 tachyzoites, RH strain Reduced tachyzoites load in liver and brain (statistically significant, P < 0.05)
TgRACK-1e Protein (35 μg), in BALB/c 4 × 104 tachyzoites, RH strain Increased survival rate of 45% relative to control [20]
Decreased parasite burden in liver andbrain (50%)

SAG, surface antigen; ROP, rhoptry antigen; GRA, dense granule antigen; IFA, incomplete Freund’s adjuvant; FCA, Freund’s complete adjuvant; CpG, oligodeoxynucleotides CpG motif adjuvant; sc, subcutaneous; im, intramuscular; ip, intraperitoneal; in, intranasal.

a Antigen encapsulation in poly (lactide-co-glycolide) microparticles as adjuvant and sustained-release of antigen.

b Mice were fed with 83 mg of leaf extracts, containing an equivalent of 1 μg of rGRA4 in 5 doses at 1 week intervals.

c T. gondii actin.

d T. gondii protein disulfide isomerase (PDI), localized mainly in the ER, involved in cell signaling and homeostasis; also found on tachyzoite membrane surface and modulates host-parasite cell interactions.

e T. gondii receptor for activated C kinase-1.

Table 2.
Examples of immunization with DNA vaccines against Toxoplasma gondii
Antigen Antigen delivery Mouse strain Challenge Results Study
TgPLP1a Plasmid, im Kunming 1 × 103 tachyzoites, RH strain Increased survival time (11.3 ± 0.9 days). Unvaccinated cohort succumbed by day 6 [59]
TgPLP1 Plasmid+IL-18, im Kunming 1 × 103 tachyzoites, RH strain Improved survival time with IL18 co-administration (12.7 ± 1.2 days) [59]
ROP9 Plasmids, im Kunming 1 × 103 tachyzoites, RH strain Increased survival time (12.9 ± 2.9 days) [35]
ROP8 Plasmid, im BALB/c 1 × 103 tachyzoites, RH strain Increased survival rate (50%)b [52]
ROP13 Plasmid+IL-18, im Kunming Acute: 1 × 103 tachyzoites (RH) RH strain challenge: Increased survival time (32.3 ± 2.7 days) [55]
Chronic: 10 tissue cysts, PRU (Type II) strain PRU strain challenge: Reduction in brain tissue cysts load (66%)
ROP16 Plasmid, im Kunming 1 × 103 tachyzoites, RH strain Improved survival time (21.6 ± 9.9 days) [56]
ROP18 Plasmid, im Kunming 1 × 103 tachyzoites, RH strain Increased survival time (27.9 ± 15.1 days). Unvaccinated cohort succumbed by day 7 [57]
SAG1 Plasmid, im BALB/c 1 × 103 tachyzoites, RH strain No complete protection, 100% mortality by day 8 [46]
SAG1+SAG3 Plasmids, im BALB/c 1 × 103 tachyzoites, RH strain Increased survival rate (20%) [46]
SAG1+SAG3 Plasmids+CT, im BALB/c 1 × 103 tachyzoites, RH strain Increased survival rate with CT adjuvant (40%). Increased secretion of IFN-γ and higher lymphocyte proliferation [46]
SAG1 Plasmid, im BALB/c 1 × 104 tachyzoites, RH strain Increased survival time (7.7 ± 2.5 days) [47]
SAG1+14-3-3c Plasmids, im BALB/c 1 × 104 tachyzoites, RH strain Increased survival time (11.5 ± 3.5 days) [47]
SAG2C+SAG2D+SAG2Xd Plasmids, im BALB/c 20 tissue cysts, PRU (Type II) strain Significant reduction in brain tissue cysts: SAG2C (72%), SAG2X (69%) and combined SAG2CDX (77%) [61]
No significant reduction in brain cysts for SAG2D-immunized group
SAG1+GRA2 Plasmids, im BALB/c 1 × 104 tachyzoites, RH strain All mice succumbed by day 16 (n = 12) [48]
SAG1+GRA2 Plasmids+pSPreS2e, im BALB/c 1 × 104 tachyzoites, RH strain All mice succumbed by day 17 (n = 12) [48]
No significant difference between immunization with or without pSPreS2 adjuvant
AMA1 Plasmid+recombinant adenovirus, ip C57BL/6 1 × 103 tachyzoites, PLK-GFP (Type II) strain Heterologous prime-boost strategy using plasmid and virus vector bearing AMA1 antigen showed 50% survival rate, 23% reduction in brain tissue cysts [62]
GRA4 Plasmid+peptide, im BALB/c 1 × 103 tachyzoites, RH strain Heterologous prime-boost strategy using plasmid and GRA4 peptide antigen showed increased survival time (16.5 ± 5.4 days) and 40% survival rate [63]
GRA6f Plasmid+LMSg, im BALB/c 1 × 103 tachyzoites, RH strain Partial protection. Higher survival rate with LMS (53.3%). Survival rate without adjuvant was lower (40%). Control group had 0% survival [54]
No detectable parasites in brain, liver and spleen of immunized groups (with and without LMS)
MIC13 Plasmid, im Kunming Acute: 1 × 103 tachyzoites, RH strain RH strain challenge: Increased survival time (21.3 ± 11.3 days). [34]
Chronic: 10 tissue cysts, PRU (Type II) strain PRU strain challenge: Reduction in brain tissue cysts load (57%)
MIC8 Plasmid+IL-12, im Kunming 1 × 103 tachyzoites, RH strain Increased survival time (15 days). No complete protection, all mice succumbed to illness [64]
MIC8 Plasmid+mIL-21+mIL-15, im Kunming Acute: 1 × 103 tachyzoites, RH strain RH strain challenge: Increased survival time (16.2 ± 1.3 days) [43]
Chronic: 20 tissue cysts, PRU (Type II) strain PRU strain challenge: Reduction in brain tissue cysts load (63.8%)
TgCDPK5h Plasmid, im Kunming Acute: 1 × 103 tachyzoites, RH strain Increased survival time (8.7 ± 4.3 days) [33]
Chronic: 10 tissue cysts, PRU strain Reduced brain tissue cysts (40%)
TgCDPK3i Plasmid, im Kunming Acute: 1 × 103 tachyzoites, RH strain Increased survival time (13.5 ± 4.9 days) [60]
Chronic: 10 tissue cysts, PRU strain Reduced brain tissue cysts (54%)
TgCyPj Plasmid, im BALB/c 500 tachyzoites, RH strain Increased survival rate (37.5%) [53]
No reduction in brain parasite load
eIF4Ak Plasmid, im Kunming 1 × 103 tachyzoites, RH strain Increased survival time (23.0 ± 5.5 days) [36]
eIF2α Plasmid, im Kunming Acute: 1 × 103 tachyzoites, RH strain Increased survival time (15.9 ± 4.6 days) [65]
Chronic: 10 tissue cysts, PRU (Type II) strain Reduced brain tissue cysts (44%)
TgROM1l Plasmid, im BALB/c 1 × 103 tachyzoites, RH strain Increased survival time (12.5 ± 0.7 days) [58]
TgIMP-1m Plasmid, im BALB/c 500 tachyzoites, RH strain Increased survival time (15.8 ± 6 days) [66]

SAG, surface antigen; ROP, rhoptry antigen; GRA, dense granule antigen; AMA, apical membrane antigen; MIC, microneme antigen; CT, cholera toxin; im, intramuscular; ip, intraperitoneal; in, intranasal.

a T. gondii perforin-like protein 1.

b Authors did not state the survival rate at endpoint of day 29, therefore the rate was inferred from the survival curve. The study showed a 100% survival at day 9 while control group had total mortality.

c Antigen 14-3-3 is a protein found on parasitophorous vacuoles (PV) of tachyzoites and is a member of excretory secretory antigen (ESA) fraction.

d SAG2C/D/X are bradyzoite-specific antigens, important in chronic infection.

e PSPreS2 is a novel genetic adjuvant encoding for hepatitis B virus surface antigen S and PreS2 - a highly immunogenic region within native HbsAg.

f DNA vaccine encodes for the immunodominant GRA6 epitope HF10 from a Type II T. gondii strain which binds the H-2LD MHC class 1 molecule.

g Levamisole (LMS), a chemical adjuvant.

h T. gondii calcium-dependent protein kinase 5.

i T. gondii calcium-dependent protein kinase 3.

j T. gondii cyclophilin.

k Eukaryotic translation initiation factor, an RNA helicase.

l Rhomboid protein 1, an intramembrane serine protease.

m T. gondii immune-mapped protein-1, a highly conserved antigen in apicomplexan parasites.

Table 3.
Examples of immunization with live, attenuated vectors expressing Toxoplasma gondii antigens
Antigen Antigen delivery Mouse strain Challenge Results Study
GRA4 DNA plasmid, gg+Vaccinia C57BL/6 2x104 tachyzoites, PLK/GFP (Type II) strair 100% survival rate. No detectable brain tissue cysts in vaccinated cohort, while controls showed 400-600 tissue cysts per brain. Significantly reduced brain tachyzoites load [67]
SAG1 Heterologous vaccination: Adenovirus, sc+MVAa, im C57BL/6 10 tissue cysts, Me49 (Type II) strain 80% survival rate, which is higher than homologous vaccination protocol (60%). Significantly reduced brain tissue cysts compared to homologous vaccination [72]
SAG1+SAG2 Salmonella typhimurium+ CT, ig BALB/c 1x103 tachyzoites, RH strain 40% survival rate [73]
SAG1 Recombinant Pseudorabies virus (rPRV), im BALB/c 50 tachyzoites, RH strain 60% survival rate with a single dose immunization [74]
Survival rate was reduced to 20% when 3 doses of rPRV were administered in 15 days intervals
SAG1+MIC3 Recombinant Pseudorabies virus (rPRV), im BALB/c 100 tachyzoites, RH strain 66.7% survival rate [75]

SAG, surface antigen; GRA, dense granule antigen; MIC, microneme antigen; CT, cholera toxin (as an adjuvant); gg, gene gun; sc, subcutaneous; im, intramuscular; ig, intragastric.

a Modified vaccinia virus ankara.

Editorial Office
Department of Molecular Parasitology, Samsung Medical Center, School of Medicine, Sungkyunkwan University,
2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea.
Tel: +82-31-299-6251   FAX: +82-1-299-6269   E-mail: kjp.editor@gmail.com
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Copyright © 2024 by The Korean Society for Parasitology and Tropical Medicine.     Developed in M2PI