2015 Fonseca
2015 Fonseca
Abstract
Chagas disease is a zoonosis caused by Trypanosoma cruzi in which the most affected organ is the heart.
Conventional chemotherapy has a very low effectiveness; despite recent efforts, there is currently no better or more
effective treatment available. DNA vaccines provide a new alternative for both prevention and treatment of a
variety of infectious disorders, including Chagas disease. Recombinant DNA technology has allowed some vaccines
to be developed using recombinant proteins or virus-like particles capable of inducing both a humoral and cellular
specific immune response. This type of immunization has been successfully used in preclinical studies and there are
diverse models for viral, bacterial and/or parasitic diseases, allergies, tumors and other diseases. Therefore, several
research groups have been given the task of designing a DNA vaccine against experimental infection with T. cruzi.
In this review we explain what DNA vaccines are and the most recent studies that have been done to develop
them with prophylactic or therapeutic purposes against Chagas disease.
Keywords: Chagas disease, Trypanosoma cruzi, DNA vaccines
2015 Arce-Fonseca et al.; licensee BioMed Central. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain
Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article,
unless otherwise stated.
Arce-Fonseca et al. Parasites & Vectors (2015) 8:121 Page 2 of 7
and in the range of picograms to femtograms after 1 to 7 family and obtained few antibodies and CTL low activity.
days of intravenous administration of DNA complexes Protection against lethal challenge was 30% to 60%. When
with cationic lipids [23]. Because the plasmids do not rep- the interleukin-12 (IL-12) and granulocyte macrophage
licate in mammalian cells, the amount available for the colony-stimulating factor (GM-CSF) genes were co-
gene expression is very low. Therefore, a strong promoter/ administered CTL activity, antibody production and re-
terminator to drive the expression of the gene must be sistance to infection with the parasite were increased.
chosen [9]. Several studies have shown that the amount of Lower parasitism in the tissues analyzed, less swelling
antigen produced after inoculation of the DNA is in the and less damage in skeletal muscle were also observed.
range of picograms to nanograms in vivo. Plasmid DNA is Survival to the challenge was 60% to 80%; greater pro-
usually inoculated into the host or animal model by direct tection by the mixture of the three genes was not gen-
injection into the muscle but there are other inoculation erated [29].These results show that trans-sialidases,
routes. The nucleic acid is taken up and expressed by the remain good candidates for vaccines, but they are a
host cells, causing the subsequent induction of both polymorphic gene family and response may vary accord-
humoral and cellular specific immune responses [9,10,24]. ingly. Co-administration of cytokine genes enhances the
This type of immunization has been successfully used in immune response.
preclinical studies and there are diverse models for viral, The Boscardin group selected an amastigote surface
bacterial and/or parasitic diseases, allergies, tumors and protein 2 (ASP-2) related-gene of T. cruzi which is an
other diseases [24,25]. In recent years, the development of antigen recognized by antibodies and T cells from in-
vaccines against parasites has had a significant progress in fected mice and humans. Four sets of genes were ampli-
which a variety of animal models and routes of administra- fied using specific primers for the asp-2 gene from
tion have been tested [26-28]; regarding Chagas disease, amastigotes cDNA of T.cruzi Y strain and a clone with
several research groups have been given the task of design- the highest degree of identity for the gene of interest
ing a DNA vaccine against experimental infection with was selected, this gene was named clone 9. BALB/c mice
T. cruzi (Table 1). immunized with a plasmid containing clone 9 gene pro-
duced specific antibodies and CD4+ T cells secreting
DNA vaccines against Chagas disease IFN-. After trypomastigote challenge a reduction in
and increased survival in the animals was observed [30].
Trans-sialidase family and surface proteins antigens The use of parasite surface proteins, secreted proteins or
proteins that can be expressed on the infected cell sur-
Garg and Tarleton immunized C57BL/6 mice with face are also of interest to identify their possible role as
ASP-1, ASP-2 and TSA-1 genes of the trans-sialidase important immunogens for protective induction.
Table 1 DNA vaccines containing Trypanosoma cruzi genes for prophylactic use
Vector Model Immune Response Survival (%) Recombinant antigen Reference
pCMVI.UBF3/2 C57BL/6 Th1 30-60 ASP-1, ASP-2 and TSA-1 29
pcDNA3 BALB/c Th1 100 Clone 9 (ASP-2) 30
pCMV BALB/c ND 25-100 TSSA 31
C57BL/6
pcDNA3 BALB/c Th1 ND catalytic domain of a trans-sialidase 32
pcDNA3.1 C57BL/6 Th1 ND TcG1, TcG2 and TcG4 33
pBK-CMV BALB/c Th1/Th2 92 TcSP 35
pBK-CMV BALB/c Th1 75 TcSSP4 36
pBK-CMV Beagle dogs Th1 100 TcSP, TcSSP4 37
pSCREEN BALB/c Th1 ND Cruzipain 40
pcDNA3 C3H/HeN Th1 100 Cruzipain 41
pCMV C57BL/6J Th1 80 LYT1 42
Vaccinia Virus BALB/c Th1 25-50 ANYNFTLV epitope 43
pcDNA3.1 BALB/c ND 70 Tc13 44
pCMV4 BALB/c and C57BL/6 transgenic mice Th1 75 PFR2 and PFR3 45
pcDNA3.1 Holtzman rats Th1/Th2 ND CCL4/MIP-1 beta chemokine 46
ND = Not determined.
Arce-Fonseca et al. Parasites & Vectors (2015) 8:121 Page 3 of 7
Miyahira and colleagues examined the role of natural in the acute stage and with an undetectable parasite level
killer (NK) T cells against T. cruzi infection; they in- during the chronic stage. Serum and cardiac IFN-,
fected CD1d-deficient mice with blood trypomastigotes tumor necrosis factor (TNF) levels and the heart inflam-
of Tulahun strain and they found that the absence of matory infiltrate decreased in vaccinated mice during
NK cells did not increase susceptibility to infection, the development of chronic disease [33]. Together, these
comparison of the parasitaemia and survival of these results demonstrated the identification of new vaccine
animals with normal BALB/c and C57BL mice being candidates that provide protection against T. cruzi in ex-
very similar. Subsequently, they analyzed whether ad- perimental mice.
ministration of -galactosylceramide (-GalCer) con- Recently, the Carlos Slim Health Institute proposed an
ferred protection, resulting in a change in parasitaemia important initiative to accelerate the development of a
only in the late stage of infection, and a slight improve- new bivalent vaccine comprising two T. cruzi recombinant
ment in survival rate when mice were infected intraperito- antigens, Tc24 and TSA-1, against Chagas disease in
neally. The combined use of -GalCer and benznidazole Mexico. A consortium including the Centro de Investiga-
did not increase the therapeutic efficacy of the drug, sug- cin y de Estudios Avanzados of the Instituto Politcnico
gesting that NK T cells do not play a major role in resist- Nacional (CINVESTAV-IPN), Laboratorios de Biolgicos
ance to infection. In order to determine whether NKcells y Reactivos de Mxico, S. A. de C. V. (BIRMEX) in Mexico
involved in the immunity induced by DNA immunization, City, and the Centro de Investigaciones Regionales Dr.
animals were immunized with the pTSSA-plasmid, which Hideyo Noguchi in Mrida, state of Yucatn was formed.
encodes a T. cruzi trans-sialidase. It was found that para- This initiative will work together with a focus on develop-
sitaemia was suppressed and survival in infected animals ing an optimal vaccine in the least possible time [34].
consequently increased in those that were plasmid- The Rosales-Encina group immunized BALB/c mice with
immunized, in comparison to animals immunized with the the TcSP gene encoding a member of the trans-sialidase
vector alone. Co-administration of -GalCer and pTSSA- family, TcSP4 gene or the recombinant proteins. The serum
plasmid immunization impaired the protective effect in- cytokine analysis showed that immunization with the re-
duced by theDNA, as the parasiatemia was increased and combinant protein or with TcSP resulted in a T cell mixed
did not improve survival compared to mice immunized Th1-Th2 response. IFN- was detected in the TcSP4 gene
only with the empty plasmid. When mice were immunized vaccinated-mice sera shortly after immunization, suggest-
with DNA encoding a CD8+ T cells epitope and -GalCer ing a Th1 response. The immunized mice were infected
was co-administered they showed a much lower CD8+ with the T. cruzi H8 strain blood trypomastigotes. Only
T cell production than those animals that were only im- mice immunized with DNA showed a significant reduction
munized [31]. This study demonstrates that the induction in the peak parasitaemia and lethal challenge survival
and activation of NK T cells can have adverse effects on [35,36]. These studies demonstrate that DNA vaccination
the protective immunity induced by DNA vaccination, induces a protective immune response in contrast to that
possibly because NK cells have Toll-like receptors capable produced by the homologous recombinant protein during
of recognizing parasite-infected cells and probably trans- T. cruzi experimental infection.
fected cells are deleted and thereby the activation of im- TcSP and TcSSP4 genes were also tested prophylactic-
mune response is decreased. ally in a canine model of Chagas disease; the antibody
Mussalem et al. used Propionibacterium acnes or a analysis revealed that the dominant subclass was IgG2.
soluble polysaccharide extracted from the bacterial wall Immunization with both recombinant plasmids induced
to modulate the immune response induced by the p154/ cell mediated immunity characterized by lymph prolifer-
13 plasmid containing a gene coding for the catalytic do- ation and IFN- production [37]. In these same animals,
main of a T. cruzi trans-sialidase. Treatment with these immunization decreased the quality and quantity of elec-
adjuvants decreased the parasitaemia peak and increased trocardiographic changes, thereby preventing progres-
the Th1 specific immune response toward the trans- sion to more serious cardiac disorders [38]. A partial
sialidase as was observed by a low IgG1/IgG2a ratio and protective effect for the prevention of macroscopic and
in vitro strong synthesis of IFN- by CD4 T cells [32]. microscopic damage in cardiac tissue during the chronic
These data suggest that adjuvants can improve the DNA phase was also observed [39]. Two T. cruzi genes gener-
vaccines protection against T. cruzi. ating a moderate level of protection in the chronic phase
Bathia and Garg vaccined C57BL/6 mice with three of the disease were proposed.
doses of DNA plasmid encoding TcG1, TcG2 and TcG4
antigens which are expressed in the plasma membrane Antigenic proteins
of trypomastigotes/amastigotes and also with plasmids
encoding IL-12 and GM-CSF. IgG2b/IgG1 isotypes were Another T. cruzi antigen that has been studied by re-
associated with 50%-90% of tissue parasite load control searchers is cruzipain (Cz). It has been shown that this
Arce-Fonseca et al. Parasites & Vectors (2015) 8:121 Page 4 of 7
protein is antigenic both in humans and in mice during individual genes and multigene families can serve as ef-
parasite infection. Schanpp et al. immunized mice with fective vaccines.
the Cz gene and showed a CTL response capable of rec- The Miyahira group used a recombinant adenovirus
ognizing and lysing cells infected with T. cruzi [40]. and the vaccinia virus as vectors expressing a CD8+ T
Some antigens may be immunogenic and induce protec- cells epitope (ANYNFTLV), which is derived from a T.
tion using recombinant protein or DNA vaccination, cruzi antigen. The immunization with adenovirus and
such as the Cz gene, which is a good candidate antigen the vaccinia virus booster was effective in inducing a cel-
for the development of an anti-T. cruzi vaccine. Cz is an lular immune response and consequently protected mice
enzyme that can be secreted by the parasite and it is from parasite lethal challenge. The response was increased
considered a virulence factor so if it can be blocked or by co-administration of recombinant vaccinia virus ex-
recognized on the surface of infected cells by CTL then pressing the receptor of the nuclear factor kappa-light-
will be a good vaccine candidate, as was observed in this chain-enhancer of activated B cells (NFB) ligand as
study. adjuvant [43]. The results of this group are spectacular
Cazorla et al. evaluated mucosal immunization in C3H/ identifying an eight amino acid region of a molecule
HeN mice which received Salmonella carrying a plasmid able to induce a sufficient immune response to control
coding for the Cz (SCz) together with bacteria containing a T. cruzi lethal infection, but again keep in mind that
GM-CSF encoding plasmid. As an additional strategy, the peptides to CD4T or CD8T depend on the type of
prime-boost protocols were established in which mice were MHC for its proper recognition.
vaccinated with SCz and were subsequently boosted with Garca et al. did not find antibodies when DNA plasmid
recombinant Cz (rCz) co-administered with different adju- containing the gene of the Tc13 antigen of the T. cruzi
vants, such as CpG-ODN and MALP-2 motifs. Protocols Tulahun strain (Tc13 Tul) was administered to BALB/c
of four SCz oral doses induced a mucosal immune re- mice; however, vaccination induced specific memory T
sponse, mainly characterized by the secretion of immuno- cells with no IFN- production. From 40% to 80% of the
globulin A (IgA) and cell proliferation of gut-associated mice immunized showed signs of hepatotoxicity and heart,
lymphoid tissues, with a weak systemic immune response. liver and spleen changes five months post-immunization.
In contrast, the protocol including a boost with rCZ and After challenge, a significant reduction in parasiatemia
CpG-ODN produced a strong systemic immune response during the acute stage was observed without modification
reflected in the titres of specific IgG against Cz, splenocyte in the survival rate. The immunization resulted in de-
proliferation, interferon-gamma (IFN-) secretion, and creased severity of cardiac damage in the chronic stage
delayed hypersensitivity response. The challenge of vac- [44]. Although in this study it is reported that the chosen
cinated mice resulted in significantly lower levels of pa- antigen induces protection in the chronic stage of the dis-
rasitaemia. Parasite control was also evident from the ease, hepatotoxicity, as well as heart and spleen damage
reduction of tissue damage [41]. The results demonstrate were also demonstrated. It is very important that chosen
that administration of Salmonella-mediated Cz-DNA alone vaccination antigens are completely safe and they induce a
or in combination with CpG-ODN or rCz and MALP-2 significant level of protection.
promotes the immune response to control both the infec- Morell et al. immunized mice with DNA containing
tion with T. cruzi and the collateral damage to muscle genes forT. cruzi paraflagelar rod proteins (PFR2 and PFR3)
tissue. alone or fused to the 70 kilodalton heat shock protein
Fralish and Tarleton performed the screening of a (HSP70). Both immunizations induced high levels of anti-
cDNA expression library of T. cruzi amastigote with PFRs IgG2a; however, only the PFR2-HSP70 immunization
monoclonal anti-amastigote antibodies and identified induced a significant increase in the IL-12 and IFN- ex-
the FCaBP gene encoding the flagellar Ca(2+) binding pression and a decrease in the percentage of cells express-
protein, Tc3 gene encoding a new homologue of the ing IL-4 [45]. The chimerical gene immunization may
adaptin AP-3 complex 3 subunit and LYT1 gene which provide a protective response against T. cruzi experimental
encodes a secreted T. cruzi protein involved in cell lysis infection.
and infectivity. The three gene peptides induced a re-
sponse of cytotoxic T cells in chronically infected mice; Non-specific T. cruzi proteins
however, the immunization with only LYT1 gene pro-
tected 80% of mice from T. cruzi lethal challenge. Alter- Roffe et al. injected a cardio-toxin and four doses of
natively gene mixtures were tested in immunization 100 g of plasmid encoding for the chemokine (C-C
assays [42]. This study demonstrates that the ability of motif ) ligand 4/macrophage inflammatory protein 1 beta
T. cruzi proteins to induce immune responses in in- (CCL4/MIP-1 beta) into rats intramuscularly. After 14
fected hosts, is not necessarily associated with the cap- days after the last immunization the rats were chal-
acity to induce protection and thus the products of the lenged with T. cruzi CL-Brener clone. The myocarditis
Arce-Fonseca et al. Parasites & Vectors (2015) 8:121 Page 5 of 7
was still intense at day 30 but the inflammatory infiltrate treatment for the chronic phase. The effects of these
showed a focal distribution. Increased anti-CCL4/MIP- changes in the infection control required long periods of
1beta levels in the T. cruzi infected animals were in- time to be detectable but resulted in a myocarditis and
duced by the immunization. This was associated with an parasite load reduction in both stages of the infection as
exacerbation of inflammation and fibrosis of the myocar- tested by histopathological analysis and PCR semi-
dium, although no changes were observed in the myo- quantitative detection of T.cruzi in cardiac tissue [49].
cardial parasitism and parasitaemia rate [46]. This study These results suggest that the DNA vaccines inducing
suggests that CCL4/MIP-1 beta plays a role in prevent- CD8+ T cell activity and IFN- production, as Tc24 gene,
ing excessive inflammation rather than the control of might be good candidates for effective therapeutic vaccin-
parasite replication. ation against T. cruzi infection.
Aparicio-Burgos et al. induced an antigen specific IgM
DNA vaccines as immunotherapy against Chagas disease and IgG response (IgG2a, IgG1) in dogs after vaccin-
All antigens that have been used in the formulation of ation with TcVac1, which consisted of various plasmids
therapeutic DNA vaccines belong to the trans-sialidase encoding T. cruzi antigens, IL-12 and GM-CSF. In-
family. creased CD8+ T cell proliferation and IFN- production
The Sanchez-Burgos group performed intraperitoneal as well as suppression of phagocyte activity, were ob-
infection in ICR mice with 500 parasites. Five and 12 days served. Parasitaemia was controlled and a moderate de-
later mice were treated with 20 g of DNA plasmid con- crease in the triatomine infectivity was produced by the
taining theTSA-1, TS, ASP-2-like, Tc52 and Tc24 antigens. vaccination; however, heart parasite load and electrocar-
The DNA treatment with the gene encoding Tc52 reduced diographic and histopatologic alterations were not pre-
the parasitaemia and heart parasite load and improved the vented but were less severe than in non-immunized
survival, but myocarditis was not affected significantly. dogs [50]. These results showed a moderate effect on
The plasmids encoding Tc24 and TSA-1 significantly re- both the acute and the chronic stages of infection but do
duced parasitaemia, mortality, myocarditis and heart para- not avoid the disease.
site load [47]. These data demonstrate that the therapeutic Rigatos group used a plasmid and a human adenovirus
vaccination efficacy is dependent on the antigen and sug- Type 5 (HuAd5) with a replication defect expressing the
gest that DNA vaccines encoding Tc24, TSA-1 and Tc52 T. cruzi amastigote surface protein 2 (ASP-2). The aim
represent good candidates for further studies for a thera- was to elucidate the immmuno-protective memory T cell
peutic vaccine against Chagas disease. phenotype and function. Short and long-term CD8+ T
Dumonteil et al. infected BALB/c mice with lethal cell populations available were compared in detail after
doses of parasites and subsequently mice were treated immunization. It was found that despite the timing, both
with two immunizations of 100 g of DNA plasmid populations overlapped greatly with respect to functional
containing the Tc24 antigen, starting on day 5 post- and phenotypic characteristics [51]. In a subsequent study
infection. The treated animals showed parasitaemia and it was observed that the up-regulation of CD95 expression
heart tissue inflammation reduction; the survival was and the phenotype of the pro-apoptotic pathogen-specific
more than 70% with TSA-1 and 100% with Tc24. Para- CD8+ T cells expanded during infection were significantly
sitological heart tissue analysis indicated that most of reduced when the adenoviral vaccine was provided at the
the mice contained parasites kinetoplast DNA but a few time of the infection. In parallel, mice vaccinated with
of them showed living parasites, suggesting that the im- adenovirus and subsequently infected had a stronger cell
munotherapy was effective but did not totally eliminate immune response mediated by CD8 T cells and survived
parasites [48]. In this strategy where the infection is an otherwise lethal infection. It was concluded that a
already established, the use of therapeutic vaccines may suboptimal response of CD8+ T cells is associated with
help to reduce the risk of developing the acute stage of up-regulation of CD95 expression and a pro-apoptotic
infection and therefore the associated pathology, but the phenotype. Both situations can be blocked by the adeno-
parasite cannot be totally eliminated. viral vaccination [52].
In a subsequent study, Zapata et al. investigated changes Hoft et al. tested whether co-administration of a plas-
of T cell populations induced by DNA vaccine as immuno- mid encoding IL-15 (pIL-15) and a plasmid encoding
therapy. The ICR mice were infected with 500 blood try- trans-sialidase (pTS) of Trypanosoma cruzi could im-
pomastigotes and treated during the acute or chronic prove the duration of protection, this co-administration
stage with two doses of 100 g of DNA. Flow cytometry did not significantly alter the T cell response or the pro-
indicated that the therapeutic vaccine induced a rapid tection shortly after immunization. However, mice vacci-
increase in the number of CD4+ and CD8+ T cells in both nated with both plasmids and challenged 6 months post
the acute and chronic stages. Also there was a rapid immunization were significantly more protected than
increase in IFN- production by CD8+ T cells after those who received only the gene of T. cruzi. Improved
Arce-Fonseca et al. Parasites & Vectors (2015) 8:121 Page 6 of 7
protection correlated with a significant increase of spe- coding for cruzipain; rCz: recombinant cruzipain; Ig: Immunoglobulin;
cific IFN-producing T cells against trans-sialidase [53]. IFN-: Interferon-gamma; ASP: Amastigote surface protein; NK: Natural killer;
-GalCer: -galactosylceramide; FCaBP: Flagellar Ca(2+) binding protein;
It was shown here that IL-15 may have a significant ef- Tc3: Homologue of the adaptin AP-3 complex 3 subunit; NFB: Nuclear factor
fect as an adjuvant for induction of increased protection kappa-light-chain-enhancer of activated B cells; MHC: Major histocompatibility
against T. cruzi. complex; PFR: Paraflagelar rod protein; HSP70: 70 kilodalton heat shock protein;
CCL4/MIP-1 beta: Chemokine (C-C mofif) ligand 4/macrophage inflammatory
Nogueira et al. used the yellow fever virus as a vector protein 1 beta; TNF: Tumor necrosis factor; CINVESTAV-IPN: Centro de
to immunize mice with the gene encoding the T. cruzi Investigacin y de Estudios Avanzados of the Instituto Politcnico Nacional;
ASP-2. It was shown that the recombinant virus YF17D/ BIRMEX: Laboratorios de Biolgicos y Reactivos de Mxico, S. A. de C. V.;
HuAd5: Human adenovirus type 5.
ENS1/Tc was capable of priming CD8+ T cells directed
against the T. cruzi TEWETG1 epitope producing IFN-
Competing interests
after challenge [54]. This study shows that the use of The authors declare that they have no competing interests.
viral formulations can be a good strategy to induce pro-
tective immune responses against pathogens. Authors contributions
MAF conceived and designed the study; MRC, SCCS and MCM drafted the
DNA vaccines, DNA vaccines against Chagas disease and DNA vaccines as
Conclusions immunotherapy against Chagas disease sections, respectively; ORM performed
DNA vaccines have provent o be effective in a variety of critical review of the contents of the entire article. MAF and ORM wrote the
preclinical models. This technology has been adopted by paper. All authors read and approved the final version of the review manuscript.
many researchers as a strategy to develop a suitable Received: 1 October 2014 Accepted: 13 February 2015
vaccine against Chagas disease. It has explored various im-
munogens (genes encoding a variety of T. cruzi proteins),
and routes of administration as well as the use of References
immunomodulators. 1. World Health Organization. Weekly epidemiological record. Geneve,
Switzerland 2012;87:509-26. Available from: http://www.who.int/wer
Although no vaccine has been able to prevent the in- 2. Viotti R, Vigliano C, Armenti H, Segura E. Treatment of chronic Chagas'
fection, in some cases immunizations have induced par- disease with benznidazole: clinical and serologic evolution of patients with
tial protection; these studies provide data that proves the long-term follow-up. Am Heart J. 1994;127:15162.
3. Murcia L, Carrilero B, Segovia M. Limitations of currently available Chagas
feasibility of preventive and therapeutic DNA vaccines to disease chemotherapy. Rev Esp Quimioter. 2012;25:13.
control the T. cruzi infection. However, a better under- 4. Astelbauer F, Walochnik J. Antiprotozoal compounds: state of the art and
standing of the protective immune responses that can ef- new developments. Int J Antimicrob Agents. 2011;38:11824.
5. Urbina JA, Docampo R. Specific chemotherapy of Chagas disease:
fectively stop parasite development and contribute to controversies and advances. Trends Parasitol. 2003;19:495501.
the understanding and development of an effective vac- 6. Dumonteil E. DNA vaccines against protozoan parasites: opportunities and
cine remains necessary. challenges. J Biomed Biotechnol. 2007;2007:90520.
7. Smirlis D, Soares MB. Selection of molecular targets for drug development
Prophylactic vaccines in humans would be a very ef- against trypanosomatids. Subcell Biochem. 2014;74:4376.
fective control method for Chagas disease, which would 8. Soeiro MN, Werbovetz K, Boykin DW, Wilson WD, Wang MZ, Hemphill A.
also be further improved if they were also used in the Novel amidines and analogues as promising agents against intracellular
parasites: a systematic review. Parasitology. 2013;140:92951.
veterinary sectors. 9. Montgomery DL, Ulmer JB, Donnelly JJ, Liu MA. DNA Vaccines. Pharmacol
The therapeutic use of DNA vaccines against Chagas Ther. 1997;74:195205.
disease could improve the effectiveness of current treat- 10. Coban C, Kobiyama K, Aoshi T, Takeshita F, Horii T, Akira S, et al. Novel
strategies to improve DNA vaccine immunogenicity. Curr Gene Ther.
ment and thus provide a better prognosis for the patient. 2011;11:47984.
These vaccines for therapeutic use have shown no ad- 11. Prompetchara E, Ketloy C, Keelapang P, Sittisombut N, Ruxrungtham K.
verse or toxic effects. Induction of Neutralizing Antibody Response against Four Dengue Viruses
in Mice by Intramuscular Electroporation of Tetravalent DNA Vaccines.
The prophylactic or therapeutic DNA vaccines have PLoS ONE. 2014;9:e92643.
been shown to reduce the duration and signs of the 12. Dutton JL, Li B, Woo WP, Marshak JO, Xu Y, Huang ML, et al. A novel DNA
acute stage, whereas in the chronic phase have avoided vaccine technology conveying protection against a lethal herpes simplex
viral challenge in mice. PLoS ONE. 2013;8:e76407.
or lessened the severity of heart damage and thereby 13. Obeng-Adjei N, Hutnick NA, Yan J, Chu JS, Myles DJ, Morrow MP, et al.
prolonged the patients lifetime. DNA vaccine cocktail expressing genotype A and C HBV surface and
Prophylactic vaccines have failed so far to prevent consensus core antigens generates robust cytotoxic and antibody responses
in mice and Rhesus macaques. Cancer Gene Ther. 2013;20:65262.
parasitic infection; therefore, the current challenge is to 14. Olgasi C, Dentelli P, Rosso A, Iavello A, Togliatto G, Toto V, et al. DNA
identify the ideal antigen. vaccination against membrane-bound Kit ligand: a new approach to
There is a need to create new formulations that can inhibiting tumour growth and angiogenesis. Eur J Cancer. 2014;50:23446.
15. Liu H, Geng S, Feng C, Xie X, Wu B, Chen X, et al. A DNA vaccine targeting
improve the existing protection achieved so far, as well p42.3 induces protective antitumor immunity via eliciting cytotoxic CD8+T
as to test new methods of administration. lymphocytes in a murine melanoma model. Hum Vaccin Immunother.
2013;9:2196202.
Abbreviations 16. Ho PP, Fontoura P, Ruiz PJ, Steinman L, Garren H. An immunomodulatory
CTL: Cytotoxic T lymphocytes; IL: Interleukin; GM-CSF: Granulocyte-macrophage GpG oligonucleotide for the treatment of autoimmunity via the innate and
colony-stimulating factor; Cz: Cruzipain; SCz: Salmonellacarrying a plasmid adaptive immune systems. J Immunol. 2003;171:49206.
Arce-Fonseca et al. Parasites & Vectors (2015) 8:121 Page 7 of 7
17. Wang Y, Wang YM, Wang Y, Zheng G, Zhang GY, Zhou JJ, et al. DNA 38. Rodrguez-Morales O, Prez-Leyva MM, Ballinas-Verdugo MA, Carrillo-Snchez SC,
vaccine encoding CD40 targeted to dendritic cells in situ prevents the Rosales-Encina JL, Alejandre-Aguilar R, et al. Plasmid DNA immunization with
development of Heymann nephritis in rats. Kidney Int. 2013;83:22332. Trypanosoma cruzi genes induces cardiac and clinical protection against Chagas
18. Jgou JF, Chan P, Schouft MT, Gasque P, Vaudry H, Fontaine M. Protective disease in the canine model. Vet Res. 2012;43:79.
DNA vaccination against myelin oligodendrocyte glycoprotein is overcome 39. Rodrguez-Morales O, Carrillo-Snchez SC, Garca-Mendoza H, Aranda-Fraustro A,
by C3d in experimental autoimmune encephalomyelitis. Mol Immunol. Ballinas-Verdugo MA, Alejandre-Aguilar R, et al. Effect of the plasmid-DNA
2007;44:3691701. vaccination on macroscopic and microscopic damage caused by the
19. Weiss R, Scheiblhofer S, Thalhamer J. Allergens are not pathogens: Why experimental chronic Trypanosoma cruzi infection in the canine model.
immunization against allergy differs from vaccination against infectious Biomed Res Int. 2013;2013:826570.
diseases. Hum Vaccin Immunother. 2014;10:7037. 40. Schnapp AR, Eickhoff CS, Scharfstein J, Hoft DF. Induction of B- and T-cell
20. Song LQ, Li Y, Li WN, Zhang W, Qi HW, Wu CG. Safety and immunogenicity respondes to cruzipain in the murine model of Trypanosoma cruzi infection.
of a DNA vaccine encoding human calcium-activated chloride channel 1 Microbes Infect. 2002;4:80513.
(hCLCA1) in asthmatic mice. Int Arch Allergy Immunol. 2013;161:24351. 41. Cazorla SI, Becker PD, Frank FM, Ebensen T, Sartori MJ, Corral RS, et al. Oral
21. Pulsawat P, Pitakpolrat P, Prompetchara E, Kaewamatawong T, vaccination with Salmonella enterica as a cruzipain-DNA delivery system
Techakriengkrai N, Sirivichayakul S, et al. Optimization of a Der p 2-based confers protective immunity against Trypanosoma cruzi. Infect Immun.
prophylactic DNA vaccine against house dust mite allergy. Immunol Lett. 2008;76:32433.
2013;151:2330. 42. Fralish BH, Tarleton RL. Genetic immunization with LYT1 or a pool of
22. Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, et al. Direct trans-sialidase genes protects mice from lethal Trypanosoma cruzi infection.
gene transfer into mouse muscle in vivo. Science. 1990;247:14658. Vaccine. 2003;21:307080.
23. Lew D, Parker SE, Latimer T, Abai AM, Kuwahara-Rundell A, Doh SG, et al. 43. Miyahira T, Takashima Y, Kobayashi S, Matsumoto Y, Takeuchi T, Ohyanagi-Hara
Cancer gene therapy using plasmid DNA: pharmacokinetic study of DNA M, et al. Immune response against a single CD8+ -T cell epitope induced by virus
following injection in mice. Hum Gene Ther. 1995;6:55364. vector vaccination can successfully control Trypanosoma cruzi infection. Infect
24. Waine GJ, McManus DP. Nucleic acids: vaccines of the future. Parasitol Immun. 2005;73:735665.
Today. 1995;11:1136. 44. Garca GA, Arnaiz MR, Laucella SA, Esteva MI, Ainciart N, Riarte A, et al.
25. Mairhofer J, Lara AR. Advances in host and vector development for the Immunological and pathological responses in BALB/c mice induced by
production of plasmid DNA vaccines. Methods Mol Biol. 2014;1139:50541. genetic administration of Tc 13 antigen of Trypanosoma cruzi. Parasitology.
26. Hassan IA, Wang S, Xu L, Yan R, Song X, Li X. DNA vaccination with a gene 2006;132:85566.
encoding Toxoplasma gondii Deoxyribose Phosphate Aldolase (TgDPA) 45. Morell M, Thomas MC, Caballero T, Alonso C, Lpez MC. The genetic
induces partial protective immunity against lethal challenge in mice. immunization with paraflagellar rod protein-2 fused to the HSP70 confers
Parasit Vectors. 2014;7:431. protection against late Trypanosoma cruzi infection. Vaccine. 2006;24:704655.
27. Cong H, Zhang M, Xin Q, Wang Z, Li Y, Zhao Q, et al. Compound DNA 46. Roffe E, Souza AL, Caetano BC, Machado PP, Barcelos LS, Russo RC, et al. A
vaccine encoding SAG1/ SAG3 with A2/B subunit of cholera toxin as a DNA vaccine encoding CCL4/MIP-1beta enhances myocarditis in
geneticadjuvant protects BALB/c mice against Toxoplasma gondii. experimental Trypanosoma cruzi infection in rats. Microbes Infect.
Parasit Vectors. 2013;6:63. 2006;8:274555.
28. Wang X, Zhang L, Chi Y, Hoellwarth J, Zhou S, Wen X, et al. The nature and 47. Sanchez-Burgos G, Mezquita-Vega RG, Escobedo-Ortegon J, Ramirez-Sierra
combination of subunits used in epitope-based Schistosoma japonicum MJ, Arjona-Torres A, Ouaissi A, et al. Comparative evalutation of therapeutic
vaccine formulations affect their efficacy. Parasit Vectors. 2010;3:109. DNA vaccines against Trypanosoma cruzi in mice. FEMS Immunol Med
29. Garg N, Tarleton R. Genetic immunization elicits antigen-specific protective Microbiol. 2007;50:33341.
immune responses and decreases disease severity in Trypanosoma cruzi 48. Dumonteil E, Escobedo-Ortegon J, Reyes-Rodriguez N, Arjona-Torres A,
infection. Infect Immun. 2002;70:554755. Ramirez-Sierra MJ. Immunotherapy of Trypanosoma cruzi infection with DNA
vaccines in mice. Infect Immun. 2004;72:4653.
30. Boscardin SB, Kinoshita SS, Fujimura AE, Rodrigues MM. Immunization with
49. Zapata-Estrella H, Hummel-Newell C, Sanchez-Burgos G, Escobedo-Ortegon J,
cDNA expressed by amastigotes of Trypanosoma cruzi elicits protective
Ramirez-Sierra MJ, Arjona-Torres A, et al. Control of Trypanosoma cruzi
immune response against experimental infection. Infect Immun.
infection and changes in T-cell populations induced by a therapeutic DNA
2003;71:274457.
vaccine in mice. Immunol Lett. 2006;103:18691.
31. Miyahira Y, Katae M, Takeda K, Yagita H, Okumura K, Kobayashi S, et al.
50. Aparicio-Burgos JE, Ochoa-Garca L, Zepeda-Escobar JA, Gupta S, Dhiman M,
Activation of natural killer T cells by alpha-galactosylceramide impairs DNA
Martnez JS, et al. Testing the efficacy of a multi-component DNA-prime/
vaccine-induced protective immunity against Trypanosoma cruzi.
DNA-boost vaccine against Trypanosoma cruzi infection in dogs. PLoS Negl
Infect Immun. 2003;71:123441.
Trop Dis. 2011;5:e1050.
32. Mussalem JS, Vasconcelos JR, Squaiella CC, Ananias RZ, Braga EG, Rodrigues
51. Rigato PO, de Alencar BC, de Vasconcelos JR, Dominguez MR, Arajo AF,
MM, et al. Adjuvant effect of the Propionibacterium acnes and its purified
Machado AV, et al. Heterologous plasmid DNA prime-recombinant human
soluble polysaccharide on the immunization with plasmidial DNA
adenovirus 5 boost vaccination generates a stable pool of protective
containing a Trypanosoma cruzi gene. Microbiol Immunol. 2006;50:25363.
long-lived CD8(+) T effector memory cells specific for a human parasite,
33. Bhatia V, Garg NJ. Previously Unrecognized Vaccine Candidates Control
Trypanosoma cruzi. Infect Immun. 2011;79:212030.
Trypanosoma cruzi Infection and Immunopathology in Mice. Clin Vaccine
52. Vasconcelos JR, Brua-Romero O, Arajo AF, Dominguez MR, Ersching J, de
Immunol. 2008;15:115864.
Alencar BC. Pathogen-induced proapoptotic phenotype and high CD95
34. Dumonteil E, Bottazzi ME, Zhan B, Heffernan MJ, Jones K, Valenzuela JG,
(Fas) expression accompany a suboptimal CD8+ T-cell response: reversal by
et al. Accelerating the development of a therapeutic vaccine for human
adenoviral vaccine. PLoS Pathog. 2012;8:e1002699.
Chagas disease: rationale and prospects. Expert Rev Vaccines. 2012;11:104355.
53. Eickhoff CS, Vasconcelos JR, Sullivan NL, Blazevic A, Bruna-Romero O,
35. Salgado-Jimnez B, Arce-Fonseca M, Bayln-Pacheco L, Talams-Rohana P, Rodrigues MM, et al. Co-administration of a plasmid DNA encoding IL-15
Rosales-Encina JL. Differential immune response in mice immunized with improves long-term protection of a genetic vaccine against Trypanosoma
the A, R or C domain from TcSP protein of Trypanosoma cruzi or with the cruzi. PLoS Negl Trop Dis. 2011;5:e983.
coding DNAs. Parasite Immunol. 2013;35:3241. 54. Nogueira RT, Nogueira AR, Pereira MC, Rodrigues MM, Neves PC, Galler R,
36. Arce-Fonseca M, Ramos-Ligonio A, Lpez-Monten A, Salgado-Jimnez B, et al. Recombinant yellow fever viruses elicit CD8+ T cell responses and
Talams-Rohana P, Rosales-Encina JL. A DNA vaccine encoding for TcSSP4 protective immunity against Trypanosoma cruzi. PLoS ONE. 2013;8:e59347.
induces protection against acute and chronic infection in experimental
Chagas disease. Int J Biol Sci. 2011;7:12308.
37. Arce-Fonseca M, Ballinas-Verdugo MA, Zenteno ER, Surez-Flores D,
Carrillo-Snchez SC, Alejandre-Aguilar R, et al. Specific humoral and cellular
immunity induced by Trypanosoma cruzi DNA immunization in a canine
model. Vet Res. 2013;44:15.
BioMed Central publishes under the Creative Commons Attribution License (CCAL). Under
the CCAL, authors retain copyright to the article but users are allowed to download, reprint,
distribute and /or copy articles in BioMed Central journals, as long as the original work is
properly cited.