ArticlePDF Available

Immunotherapeutic potential of Leishmania ( Leishmania ) donovani Th1 stimulatory proteins against experimental visceral leishmaniasis

Authors:

Abstract

An effective therapeutic vaccination strategy is required for controlling visceral leishmaniasis (VL), a fatal systemic disease, through boosting the immunosuppressed state in Leishmania-infected individuals, as the majority of them living in the endemic regions exhibit either subclinical or asymptomatic infection which further often develops into a full-blown disease. Previously in our laboratory, several Th1 stimulatory recombinant proteins were successfully cloned, purified and assessed for their prophylactic efficacy against Leishmania challenge. Due to their immunostimulatory property, these proteins are needed to be evaluated for their immunotherapeutic potential in Leishmania-infected hamsters. Four proteins namely, aldolase, enolase, p45 and triose phosphate isomerase were taken up to immunize animals at different doses (50, 25 and 12.5 μg/animal). Immunization with lower doses of aldolase and enolase, i.e., 25 and 12.5 μg showed a significant decline (∼60%) in parasitic load along with an enhanced cellular immune response. These findings indicate that vaccination with above -stated Th1 stimulatory proteins is an effective immunotherapeutic approach against experimental VL. However, their efficacies may further be improved in combination with known therapeutic regimens or immunomodulators.
Immunotherapeutic potential of Leishmania (Leishmania)donovani Th1
stimulatory proteins against experimental visceral leishmaniasis
Keerti
a
, Narendra K. Yadav
a
, Sumit Joshi
b
, Sneha Ratnapriya
a
, Amogh A. Sahasrabuddhe
a
,
Anuradha Dube
b,
a
Division of Molecular & Structural Biology, CSIR Central Drug Research Institute, Lucknow 226031, India
b
Division of Parasitology, CSIR Central Drug Research Institute, Lucknow 226031, India
article info
Article history:
Received 14 October 2017
Received in revised form 22 January 2018
Accepted 12 March 2018
Available online 21 March 2018
Keywords:
Visceral leishmaniasis
Th1 stimulatory proteins
Vaccination
Immunotherapeutic
Hamsters
abstract
An effective therapeutic vaccination strategy is required for controlling visceral leishmaniasis (VL), a fatal
systemic disease, through boosting the immunosuppressed state in Leishmania-infected individuals, as
the majority of them living in the endemic regions exhibit either subclinical or asymptomatic infection
which further often develops into a full-blown disease. Previously in our laboratory, several Th1
stimulatory recombinant proteins were successfully cloned, purified and assessed for their prophylactic
efficacy against Leishmania challenge. Due to their immunostimulatory property, these proteins are
needed to be evaluated for their immunotherapeutic potential in Leishmania-infected hamsters. Four pro-
teins namely, aldolase, enolase, p45 and triose phosphate isomerase were taken up to immunize animals
at different doses (50, 25 and 12.5
l
g/animal). Immunization with lower doses of aldolase and enolase,
i.e., 25 and 12.5
l
g showed a significant decline (!60%) in parasitic load along with an enhanced cellular
immune response. These findings indicate that vaccination with above -stated Th1 stimulatory proteins is
an effective immunotherapeutic approach against experimental VL. However, their efficacies may further
be improved in combination with known therapeutic regimens or immunomodulators.
!2018 Elsevier Ltd. All rights reserved.
1. Introduction
Visceral leishmaniasis (VL), described as a phlebotomine-borne
systemic infectious disease, caused by an obligate protozoan para-
site of Leishmania (Leishmania) donovani complex [1]. World Health
Organization (WHO) has reported that from the annual 50,000 to
90,000 VL cases, nearly 90% are from the underprivileged commu-
nities belonging to the regions of Indian sub-continent
(Bangladesh, Nepal, and India), Africa (Ethiopia, Sudan, and South
Sudan), and South America (Brazil) [1]. The available chemothera-
peutics for VL are quite effective, however; several concerns such
as toxicity, deleterious side effects [2], the emergence of drug resis-
tance cases [3] and variation in clinical responses due to geograph-
ical distribution [4] enforce the need for the search of alternative
treatment(s).
For the past several years, much consideration has been given to
the vaccine development program against VL wherein many of the
potential protein antigens of L. (L.) donovani have been used pro-
phylactically with varied success [5,6]. However, a commercial
human VL vaccine still remains elusive [7]. It is observed that the
population manifesting L. (L.) donovani infection with clinical
symptoms represent only a small proportion while the majority
of the individuals remain asymptomatic and act as potent threats
for the spread of this deadly disease [8,9]. Furthermore, post
kala-azar dermal leishmaniasis (PKDL), as well as Leishmania
HIV-co-infected individuals, lead to more number of treatment
failures resulting in relapse cases [10–13]. Therefore, there is a
need for a therapeutic vaccine which can curb the persistence of
disease, though it seems to be much more challenging because
active VL leads to severe immunosuppression which needs to be
boosted up. The approach of therapeutic vaccination has been suc-
cessfully reported against canine VL [14] as well as several other
chronic diseases such as Chagas disease, tuberculosis, human
papillomavirus, HIV infection and cancer [15–18].
In case of active VL, there is a prominence of Th2 type immune
response while generation of an effective Th1 type immune
response is associated with the cure of the disease [19]. Thus, those
antigens, which are capable of skewing immune response towards
Th1 type, may be envisaged as potential therapeutic vaccine
https://doi.org/10.1016/j.vaccine.2018.03.027
0264-410X/!2018 Elsevier Ltd. All rights reserved.
Corresponding author at: Division of Parasitology, CSIR-Central Drug Research
Institute, Sector 10, Janakipuram Extension, Sitapur Road, Lucknow 226 031, Uttar
Pradesh, India.
E-mail address: a_dube@cdri.res.in (A. Dube).
Vaccine 36 (2018) 2293–2299
Contents lists available at ScienceDirect
Vaccine
journal homepage: www.elsevier.com/locate/vaccine
candidates. Several recombinant proteins of L. (L.) donovani pro-
mastigote were shown to induce a Th1-biased cellular immune
response against peripheral blood mononuclear cells (PBMCs)/-
lymphocytes of treated VL patients as well as of hamsters [20].
Few of these proteins, such as Fructose-bisphosphate Aldolase
(Aldolase), 2-phospho-D-glycerate hydrolase (Enolase), triose
phosphate isomerase (TPI) and p45 showed significant prophylac-
tic efficacy in hamsters against L. (L.) donovani challenges [21–23].
Of these, aldolase, enolase and TPI, the vital glycolytic enzymes,
have been considered to be potential vaccine targets in leishmani-
asis or other infectious diseases [24–26], whereas p45, a member
of methionine aminopeptidase family, has been shown to induce
a proliferative response against Leishmania parasite in specific
T-cell lines from VL endemic Brazilian donor [27]. In this commu-
nication, we have evaluated the immunotherapeutic potential of
these proteins in Leishmania-infected hamsters.
2. Methodology
2.1. Animals and parasite
In this study, Syrian golden hamsters (Mesocricetus auratus, 7–8
weeks old) were used as a suitable experimental host. They were
maintained by Laboratory animal facility (LAF) of CSIR-CDRI under
the regulation of the institutional animal ethics committee (IAEC,
Approval No. 150/09/Para/IAEC dated 23.10.09) following the
guidelines of the committee for the purpose of control and super-
vision of experiments on animals (CPCSEA). L. (L.) donovani strain
(MHOM/IN/80/Dd8) was procured as promastigotes from
American type culture collection (ATCC, Manassas, VA, USA) and
maintained in vitro conditions following the protocol of Garg
et al. [28]. Parasite virulence was maintained by serial passaging
of amastigotes in hamsters [29].
2.2. Preparation of soluble L. (L.) donovani antigen (SLD) and
purification of recombinant proteins
SLD was prepared from metacyclic promastigotes following the
method of Gupta et al. [30], and aliquots were kept at "80 "C until
further use. All the four Th1 stimulatory proteins of L. (L.) donovani
aldolase, enolase, p45, and TPI, were cloned and purified by affin-
ity chromatography using a Ni-NTA superflow agarose matrix
beads (Qiagen, Germany) as described previously [21–23]. The pur-
ity of the proteins was assessed by SDS-PAGE, and the endotoxin
level was tested using a Limulus amoebocyte lysate test (Pierce
LAL Chromogenic Endotoxin Quantitation kit, Thermo Fisher,
USA) following the manufacturer’s instructions. These proteins
were then passed through the amicon (Centrifugal filter devices,
Millipore, USA) followed by 2–3 washings in phosphate buffered
saline (PBS) to get rid of the residual salts of elution buffer and con-
centrated to a smaller volume. Finally, the proteins were quantified
using Bradford assay [31] and stored at "80 "C until further use.
2.3. Assessment of cytotoxicity of recombinant proteins
Cytotoxic effect of each purified recombinant protein was
assessed in vitro in peritoneal macrophages harvested from naïve
hamsters stimulated with 2% starch solution for 48 h [32]. Cells
were plated at 2 #10
5
cells per well in 100 ml of complete RPMI
in 96-well cell culture plates (Nunc, Denmark) and treated with
different concentrations (100, 10, 1
l
g/ml) of rLdEno, rLdAld,
rLdp45 and rLdTPI, each in triplicates. The viability of cells was
assessed using MTT dye 3-(4, 5-dimethylthiazol-2-yl)-2, 5-
diphenyltetrazolium bromide (Calbiochem) after incubating at
37 "C in a CO
2
incubator for 72 h and absorbance was measured
at 570 nm in a SPECTRAmax PLUS 384 microplate reader (Molecu-
lar Devices, USA) [33]. Percentage viability of macrophages was
calculated in comparison to untreated ones using following
equation:
Cell viabilityð%Þ¼ Absorbance of treated cells
Absorbance of untreated cells #100
2.4. Estimation of nitric oxide in Leishmania-infected macrophages
Peritoneal macrophages, isolated from naive hamsters were
seeded as 1 #10
6
cells/ml/well in complete RPMI-1640 in 12-
well culture plates, infected with L. (L.) donovani stationary phase
promastigotes at a ratio of 1:10 for 12 h. After the incubation,
non-internalised promastigotes were removed by washing 2–3
times with incomplete RPMI-1640 at 37 "C. The infected macro-
phages were then treated for 48 h with all the four recombinant
proteins at the concentrations of 1 and 10 mg/ml, and untreated
cells were kept as control one. Nitric oxide (NO) generation was
measured in Leishmania-infected macrophages, using flow cytom-
etry (Calibur, Becton-Dickinson, USA) with fluorescent probe
DAF-2-DA (Sigma-Aldrich, USA) that allows the determination of
intracellular NO. After the treatment, the adherent cells were
scraped, washed and resuspended in PBS, stained with DAF-2DA
(2
l
M) for 30 min at 37 "C and fluorescence were acquired in Cal-
ibur using CellQuest software. Lipopolysaccharide (LPS; 10 mg/ml,
Sigma-Aldrich, USA) treated cells served as the positive control.
The data were expressed as MFI (median fluorescence intensity).
2.5. Expression of Th1 cytokine and nitric oxide synthase (NOS2) in
Leishmania-infected macrophages
The mRNA expression of Interferon-
c
(IFN-
c
), Tumor Necrosis
Factor-
a
(TNF-
a
) and NOS2 cytokines was measured through
real-time RT-PCR. Briefly, RNA from infected peritoneal macro-
phages (1 #10
6
cells/well/ml in 12-well plate) treated with or
without proteins (at 48 h post stimulation) was extracted using
RNeasy mini isolation kit (Qiagen, Germany). RNA samples were
quantified in nanodrop (Thermo Fischer Scientific, USA) and cDNA
were synthesized using High-Capacity cDNA Reverse Transcription
Kit (Applied Biosystem, USA). Real-Time RT-PCR of these cDNA
samples was performed in iQ5 Multicolor real-time PCR detection
system (Bio-Rad) using following reaction conditions: initial
denaturation at 95 "C for 2 min followed by 40 cycles, each consist-
ing of denaturation at 95 "C for 20 s, annealing at 60 "C for 20 s and
extension at 72 "C for 16 s per cycle employing various sets of pri-
mers designed with the help of beacon designer software as listed
in Table 1. For all the gene expression studies, RPL18 was used as a
reference gene [34], and mRNA expression of different cytokines
was estimated using the 2
"
DD
Ct
method [35].
2.6. Infection and immunization
A batch of 70 hamsters was divided into 14 groups comprising
five hamsters per group. Animals of all the groups except group 14
were infected intracardially with 5 #10
5
amastigotes purified
through percoll density gradient method [36]. Fifteen days post
infection (p.i.), animals belonging to groups 1–12 were adminis-
tered thrice with three doses 12.5, 25 and 50 mg of each of the
four recombinant proteins, i.e. rLdEno, rLdAld, rLdp45 and rLdTPI
intradermally at two weeks interval. Animals of groups 13 and
14 were kept as infected and uninfected control. After 15 days of
the last dose, i.e. on day 60 p.i., the animals of all the groups were
subjected to the assessment of delayed-type hypersensitivity
(DTH) response. Twenty-four hours later they were necropsied,
2294 Keerti et al. / Vaccine 36 (2018) 2293–2299
and their splenic tissue samples were collected for the assessment
of parasite burden and immunological analysis.
2.7. Measurement of DTH response
For the measurement of DTH response, both vaccinated, and
unvaccinated infected hamsters were injected intradermally with
50
l
g of SLD antigen in 0.05 ml sterile PBS in the left hind footpad
while 0.05 ml of sterile PBS only in the right hind footpad which
served as a control. After 24 h of injection, the footpad thickness
was measured with a digital Vernier Caliper. The response was
evaluated in terms of percentage increase in footpad thickness by
measuring the difference in footpad swelling between the two
footpads (with and without SLD) in each animal [30].
2.8. Assessment of parasite burden
The splenic impression smears of infected control and protein-
treated hamsters were prepared after measuring the weight of the
spleen following necropsy. Air dried smears were fixed in metha-
nol and stained with 10% Giemsa stain (Sigma-Aldrich, USA).
Amastigotes were counted per 1000 macrophage cell nuclei, and
the results were expressed in Leishman Donovan Units (LDU) cal-
culated using following formula [37,38]:
LDU ¼amastigote number per 1000 host cell nuclei
#organ weight ðin gramsÞ
2.9. RNA extraction and real-time quantitative PCR (RT-qPCR)
Total cellular RNA was isolated from the splenic tissue samples
of the hamster from all the experimental groups using the Trizol
method as per the manufacturer’s protocol (Invitrogen, USA).
RNA samples were quantified and processed for cDNA synthesis
as described above. Real-time RT-PCR was carried out in cDNA
samples for the analysis of Th1 and Th2 cytokines expression as
described earlier.
2.10. Statistical analysis
Statistical analysis was performed using GraphPad Prism 6.01
(GraphPad Software, San Diego, CA, USA). Data were analyzed by
ordinary one-way analysis of variance and Dunnett’s multiple
comparison tests. Two sets of experiments performed for the vac-
cination studies and the results were expressed as mean ± SEM
with a p-value of < 0.05 was considered significant.
3. Results
3.1. Cytotoxicity of recombinant proteins
The percentage viability of peritoneal macrophages after incu-
bation with the recombinant proteins was determined by MTT
assay. The results revealed that the cells treated with lower con-
centrations (1 and 10
l
g/ml) of either recombinant proteins or
drug, exhibited equal or higher viability in comparison to the
untreated ones. However, when stimulated with a higher concen-
tration of 100
l
g/ml of the proteins, rLdEno, and rLdAld were
found to be entirely safe, but there was a significant decline in
the percentage viability of cells treated with rLdp45 and rLdTPI.
Similarly, the treatment with a standard anti-leishmanial drug
miltefosine at the same concentration showed substantial toxicity
(p'0.0001) in comparison to untreated cells (Fig. 1).
3.2. Treatment with recombinant proteins trigger NO generation with
increased expression of Th1 type cytokines and NOS2 in Leishmania-
infected macrophages
There was a perceptible increase in NO production in peritoneal
macrophages stimulated with 10
l
g/ml of rLdEno and rLdAld after
48 h of infection as compared to their respective infected unstim-
ulated cells and the cells stimulated with other proteins, i.e. rLdp45
and rLdTPI (Fig. 2a). LPS treated cells served as a positive control
(data not shown). These observations were further supported by
Table 1
Sequences of forward and reverse primers of hamster cytokines used for real-time quantitative PCR (RT-qPCR).
S. no. Genes Direction Primer sequence Product length
1 RPL 18 Forward 5
0
AACTCCACCTTCAATCAG 3
0
96
Reverse 5
0
GATGATCCGAAAGATGAAG 3
0
2 NOS2 Forward 5
0
TGCCTTGCATCCTCATTGG 3
0
77
Reverse 5
0
GTCGCTGTTGCCAGAAACTG 3
0
3 TNF-
a
Forward 5
0
GGAGTGGCTGAGCCATCGT 3
0
131
Reverse 5
0
AGCTGGTTGTCTTTGAGAGACATG 3
0
4 IFN-
c
Forward 5
0
GCTTAGATGTCGTGAATGG 3
0
200
Reverse 5
0
GCTGCTGTTGAAGAAGTTAG 3
0
5 IL-12 Forward 5
0
AATTACTCTGGACGGTTCAC 3
0
81
Reverse 5
0
GCTACTGCTGCTCTTGAC 3
0
6 TGF-bForward 5
0
ACGGAGAAGAACTGCTGTG 3
0
178
Reverse 5
0
GGTTGTGTTGGTTGTAGAGG 3
0
7 IL-4 Forward 5
0
CCACGGAGAAAGACCTCATCTG 3
0
75
Reverse 5
0
GGGTCACCTCATGTTGGAAATAAA 3
0
8 IL-10 Forward 5
0
GTTGCCAAACCTTATCAGAAATGA 3
0
102
Reverse 5
0
TTCTGGCCCGTGGTTCTCT 3
0
Fig. 1. The percent viability of peritoneal macrophages treated with different
concentrations of recombinant proteins rLdEno, rLdAld, rLdp45, and rLdTPI as well
as antileishmanial drug miltefosine (positive control) assayed using MTT.
Significance value (*p'0.05, ** p'0.01, *** p'0.001 and **** p'0.0001) of
treated cells was calculated in respect to untreated ones. Data represents as mean ±
SEM of three independent experiments.
Keerti et al. / Vaccine 36 (2018) 2293–2299 2295
significantly higher mRNA expression of IFN-
c
and TNF-
a
along
with NOS2 in rLdEno and rLdAld treated cells as compared to
untreated infected cells (Fig. 2b, c, and d).
3.3. Hamsters immunized with recombinant proteins exhibited
enhanced DTH response
The hamsters immunized with 12.5
l
g of rLdAld and rLdTPI as
well as 25
l
g of rLdAld, rLdEno and rLdTPI exhibited a significant
increase in footpad swelling after 24 h of SLD injection as com-
pared to infected control (Fig. 3). There was no such response
observed in rLdp45 treated hamsters. The DTH response was found
to be optimum in the group treated with 12.5
l
g of the rLdAld.
However, groups treated with a higher dose of recombinant pro-
teins, i.e. 50
l
g, showed no significant response.
3.4. Immunotherapeutic efficacy of recombinant proteins
The immunotherapeutic efficacy of the recombinant proteins in
L. (L.) donovani-infected hamsters assessed on day 60 p.i. revealed a
significant decrease in parasitic load of rLdAld and rLdEno immu-
nized groups specifically at lower doses, i.e. at 12.5 and 25
l
g
(!60%; p'0.01) per animal as compared to non-immunized ham-
sters (Fig. 4). However, there was no apparent reduction in parasite
load in hamsters treated with higher doses of these proteins. Fur-
ther, in rLdp45 treated animals, though, there was inhibition of
parasite multiplication to the tune of 55% only at 25 mg/animal
(p< 0.05), there was no effect at other doses. The treatment with
rLdTPI did not show any effect in L. (L.) donovani-infected hamsters
at any dose level.
3.5. Treatment with recombinant proteins skewed Th1 type of immune
response in Leishmania-infected hamsters
Among the Th1 cytokines, there was a significant upregulation
of mRNA expression of IL-12 in hamsters treated with 12.5 mg of
rLdAld (p'0.0001) in comparison to other vaccinated groups as
well as infected control. The expression of the other cytokine,
TNF-
a
, though, found to be elevated in rLdAld and rLdEno treated
groups at doses of 12.5 and 25 mg, was not of much significance.
Fig. 2. (a) Generation of Nitric oxide (NO) in Leishmania-infected peritoneal macrophages stimulated with recombinant proteins as compared to their respective unstimulated
cells. Data represent the median fluorescence intensity (MFI) of three independent experiments and (b, c, and d) showed mRNA expression profile of NOS2 and Th1 cytokines
(relative fold change) in Leishmania-infected peritoneal macrophages treated with recombinant proteins as compared to untreated cells. Significance value (*p'0.05, ** p'
0.01, *** p'0.001 and **** p'0.0001) of treated cells was calculated in respect to untreated ones. Data represents as mean ± SE, of three independent experiments.
Fig. 3. DTH response shown as percent increase in footpad thickness to SLD antigen
on day 60 p.i. Significance values indicated the difference between the vaccinated
groups and infected group (*p'0.05, ** p'0.01, *** p'0.001 and **** p'0.0001).
Data represents as mean ± SEM of two independent experiments with similar
results.
2296 Keerti et al. / Vaccine 36 (2018) 2293–2299
Moreover, there was a noteworthy increase in the expression of
IFN-
c
in rLdAldo (25 mg, p< 0.0001) and rLdEno (12.5 and 25 mg,
p< 0.01) treated a group of hamsters, there was only a slight
increase in other treated groups. Besides, the expression of TGF-
b, a signature of Th2 cytokines was also found to be significantly
downregulated in rLdAld (12.5 mg; p'0.05) treated group and
was marginal (not significant) in rLdEno (25 mg), and rLdTPI (25
mg) treated groups. There was, however, no apparent difference
in the expression of IL-4 cytokine in all the treated groups as com-
pared to infected control. On the contrary, level of IL-10 was found
to be significantly augmented in rLdp45 and rLdTPI (p'0.0001)
immunized hamsters at a dose of 12.5 mg/animal but there was
no noticeable change in other treated groups (Fig. 5).
4. Discussion
This study assessed the immunotherapeutic efficacy of Th1
stimulatory proteins of L. (L.) donovani which were previously
reported to possess remarkable prophylactic potential against
experimental VL [21–23]. Prior to any animal experimentation
(vaccination study), the possible cytotoxicity of an immunogen
needs to be evaluated [39]. The purified recombinant proteins,
when assessed for their cytotoxicity on peritoneal macrophages,
were found to be safer for immunization. Their potential to gener-
ate an immunostimulatory response in Leishmania-infected host
cells (macrophages) was further evaluated by NO production. It
is well documented that for controlling Leishmania infection,
induction of NOS2 is essential which subsequently oxidizes L-
arginine into NO and such enzyme activity in macrophages is usu-
ally induced by the Th1 cytokines, i.e. IFN-
c
, TNF-
a
, etc. [40]. The
recombinant proteins showed a considerable increase in intracel-
lular NO production with enhanced mRNA expression of NOS2
along with IFN-
c
and TNF-
a
which indicate their ability to induce
a Th1 type immune response. These findings led us to evaluate
their immunotherapeutic efficacy in chronic L. (L.) donovani infec-
tion in hamsters.
For dosage optimization, the therapeutic immunization was ini-
tiated with three doses of each recombinant protein, i.e. 50, 25 and
12.5 mg per animal. Out of these, four proteins, rLdAld exerted opti-
mum reduction in parasite burden in hamsters at a dose of 12.5 mg/
animal followed by rLdEno at 25 mg/animal in comparison to other
recombinant protein treated groups. Other immunological param-
eters such as DTH and mRNA cytokine responses also showed sim-
ilar trends. DTH, a signature for cell-based immunity has
frequently been used as a correlate of protection in leishmaniasis
[41]. Strong DTH response observed specifically in rLdAld and
rLdEno vaccinated groups (at the doses mentioned above) signifies
towards the Th1 driven cellular response [42,43]. Additionally, the
changes in mRNA expression profile of Th1 (IL-12, IFN-
c
, and TNF-
a
) and Th2 cytokines (IL-10, IL-4 and TGF-b) was assessed in
recombinant proteins treated groups. Amongst the Th1 cytokines,
IFN-
c
plays a crucial role in the control of Leishmania infection
by inducing innate and cellular responses which might help in kill-
ing parasite by activating macrophages whereas TNF-
a
, generates
cytotoxic effect against pathogens by actively inducing the produc-
tion of reactive nitrogen intermediates either alone or with IFN-
c
[44,45]. A higher mRNA expression of these cytokines in rLdEno
and rLdAld vaccinated groups suggests their involvement in stim-
ulating the macrophages which might help in reducing the parasite
number. However, the expression of IL-12, known to be involved in
microbicidal activity against intracellular pathogens [46], was
found to be significantly higher in a lower dose of rLdAld treated
group only. On the other hand, the mRNA level of TGF-b, which
Fig. 5. Splenic mRNA cytokine (Th1/Th2) expression profile regarding relative fold change of infected and recombinant protein vaccinated groups normalized with the values
of naïve hamsters. Significance values indicate the difference between the vaccinated groups and infected group (*p'0.05, ** p'0.01, *** p'0.001 and **** p'0.0001). Data
represents as mean ± SEM, of two independent experiments.
Fig. 4. Parasite burden expressed in LDU (No. of parasites in 1000 macrophage cell
nuclei x weight of spleen in gm) evaluated on day 60 p.i. Significance values
indicate the difference between the vaccinated groups and infected group (*p'
0.05, ** p'0.01, *** p'0.001 and **** p'0.0001). Data represents as mean ± SEM,
of two independent experiments.
Keerti et al. / Vaccine 36 (2018) 2293–2299 2297
plays a major role in parasite persistence during VL infection [47],
was found to be decreased moderately in rLdEno and significantly
in rldAld treated groups indicating the skewing of Th2 type
response towards the Th1 type. However, there was no remarkable
difference in the mRNA expression of IL-4 and IL-10 cytokines
between the groups treated with above-mentioned recombinant
proteins and that of infected controls. In rLdp45 and rLdTPI treated
groups at a 12.5 mg dose, where no parasite reduction was noticed,
the mRNA expressions of IL-10 cytokine was significantly higher
while that of IFN-
c
and TNF-
a
was lower in comparison to other
treated and control groups. These findings are substantiated by
the fact that during the active disease state, VL patients showed
high levels of the IL-10 cytokine which further hinders the activity
of pro-inflammatory cytokines such as IFN-
c
and TNF-
a
that are
involved in antiparasitic activities [48].
In a nutshell, two recombinant proteins rLdEno and rLdAld of
L. (L.) donovani exerted considerable immunotherapeutic efficacy
by activating the immune status of the infected host to overcome
the immunosuppression caused by Leishmania infection and hence
could serve as potential therapeutic vaccine candidates. Efforts are
needed to further augment their immunotherapeutic efficacies in
combination with suboptimal doses of known anti-leishmanial or
immunomodulators for effective treatment of VL endemic popula-
tion. This may facilitate the process of VL elimination through the
development of therapeutic Leishmania vaccine.
Acknowledgments
We express our sincere gratitude to the Director, CSIR-CDRI for
providing necessary facilities for conducting this work. This paper
bears the CSIR-CDRI communication No 9655.
Conflict of interest
The authors declare that they have no conflict of interest.
Funding
This work was supported by the Department of Biotechnology
(DBT) [BT/PR7299/MED/29/678/2012]. CSIR India and ICMR
India provide financial assistance regarding fellowships to K., N.K.
Y., S.J., and S.R.
References
[1] WHO. Leishmaniasis. http://wwwwhoint/mediacentre/factsheets/fs375/en/;
2017.
[2] Moore EM, Lockwood DN. Treatment of visceral leishmaniasis. J Glob Infect Dis
2010;2:151–8.
[3] Freitas-Junior LH, Chatelain E, Kim HA, Siqueira-Neto JL. Visceral leishmaniasis
treatment: what do we have, what do we need and how to deliver it? Int J
Parasitol Drugs Drug Resist 2012;2:11–9.
[4] Croft SL, Olliaro P. Leishmaniasis chemotherapy–challenges and opportunities.
Clin Microbiol Infect 2011;17:1478–83.
[5] Kumar R, Engwerda C. Vaccines to prevent leishmaniasis. Clin Trans Immunol
2014;3:e13.
[6] Joshi S, Rawat K, Yadav NK, Kumar V, Siddiqi MI, Dube A. Visceral
leishmaniasis: advancements in vaccine development via classical and
molecular approaches. Front Immunol 2014;5:380.
[7] Gillespie PM, Beaumier CM, Strych U, Hayward T, Hotez PJ, Bottazzi ME. Status
of vaccine research and development of vaccines for leishmaniasis. Vaccine
2016.
[8] Sharma MC, Gupta AK, Das VN, Verma N, Kumar N, Saran R, et al. Leishmania
donovani in blood smears of asymptomatic persons. Acta Tropica
2000;76:195–6.
[9] Picado A, Dash AP, Bhattacharya S, Boelaert M. Vector control interventions for
visceral leishmaniasis elimination initiative in South Asia, 2005–2010. Indian J
Med Res 2012;136:22–31.
[10] McQuarrie S, Kasper K, Moffatt DC, Marko D, Keynan Y. Relapse of visceral
leishmaniasis in an HIV-infected patient successfully treated with a
combination of miltefosine and amphotericin B. Can J Infect Dis Med
Microbiol 2015;26:325–9.
[11] Ramesh V, Singh R, Avishek K, Verma A, Deep DK, Verma S, et al. Decline in
clinical efficacy of oral miltefosine in treatment of post Kala-azar Dermal
Leishmaniasis (PKDL) in India. PLoS Negl Trop Dis 2015;9.
[12] Bhandari V, Kulshrestha A, Deep DK, Stark O, Prajapati VK, Ramesh V, et al.
Drug susceptibility in leishmania isolates following miltefosine treatment in
cases of visceral leishmaniasis and post kala-azar dermal leishmaniasis. PLoS
Negl Trop Dis 2012;6.
[13] van Griensven J, Carrillo E, Lopez-Velez R, Lynen L, Moreno J. Leishmaniasis in
immunosuppressed individuals. Clin Microbiol Infect: Off Publ Eur Soc Clin
Microbiol Infect Dis 2014;20:286–99.
[14] Roatt BM, Aguiar-Soares RDO, Reis LES, Cardoso JMO, Mathias FAS, de Brito
RCF, et al. A vaccine therapy for canine visceral leishmaniasis promoted
significant improvement of clinical and immune status with reduction in
parasite burden. Front Immunol 2017;8.
[15] Morrow MP, Yan J, Sardesai NY. Human papillomavirus therapeutic vaccines:
targeting viral antigens as immunotherapy for precancerous disease and
cancer. Expert Rev Vaccines 2013;12:271–83.
[16] Melero I, Gaudernack G, Gerritsen W, Huber C, Parmiani G, Scholl S, et al.
Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin
Oncol 2014;11:509–24.
[17] Cardona PJ. The progress of therapeutic vaccination with regard to
tuberculosis. Front Microbiol 2016;7:1536.
[18] Barry MA, Wang Q, Jones KM, Heffernan MJ, Buhaya MH, Beaumier CM, et al. A
therapeutic nanoparticle vaccine against Trypanosoma cruzi in a BALB/c mouse
model of Chagas disease. Hum Vaccin Immunother 2016;12:976–87.
[19] Kubar J, Fragaki K. Recombinant DNA-derived leishmania proteins: from the
laboratory to the field. Lancet Infect Dis 2005;5:107–14.
[20] Joshi S, Yadav NK, Rawat K, Tripathi CD, Jaiswal AK, Khare P, et al. Comparative
analysis of cellular immune responses in treated leishmania patients and
hamsters against recombinant Th1 stimulatory proteins of Leishmania
donovani. Front Microbiol 2016;7:312.
[21] Gupta R, Kumar V, Kushawaha PK, Tripathi CP, Joshi S, Sahasrabuddhe AA, et al.
Characterization of glycolytic enzymes–rAldolase and rEnolase of Leishmania
donovani, identified as Th1 stimulatory proteins, for their immunogenicity and
immunoprophylactic efficacies against experimental visceral leishmaniasis.
PLoS One 2014;9:e86073.
[22] Gupta R, Kushawaha PK, Tripathi CD, Sundar S, Dube A. A novel recombinant
Leishmania donovani p45, a partial coding region of methionine
aminopeptidase, generates protective immunity by inducing a Th1
stimulatory response against experimental visceral leishmaniasis. Int J
Parasitol 2012;42:429–35.
[23] Kushawaha PK, Gupta R, Tripathi CD, Khare P, Jaiswal AK, Sundar S, et al.
Leishmania donovani triose phosphate isomerase: a potential vaccine target
against visceral leishmaniasis. PLoS One 2012;7:e45766.
[24] Nandan D, Tran T, Trinh E, Silverman JM, Lopez M. Identification of leishmania
fructose-1,6-bisphosphate aldolase as a novel activator of host macrophage
Src homology 2 domain containing protein tyrosine phosphatase SHP-1.
Biochem Biophys Res Commun 2007;364:601–7.
[25] Avilan L, Gualdron-Lopez M, Quinones W, Gonzalez-Gonzalez L, Hannaert V,
Michels PA, et al. Enolase: a key player in the metabolism and a probable
virulence factor of trypanosomatid parasites-perspectives for its use as a
therapeutic target. Enzyme Res 2011;2011:932549.
[26] Zhu Y, Si J, Harn DA, Yu C, Liang Y, Ren J, et al. The protective immunity of a
DNA vaccine encoding Schistosoma japonicum Chinese strain triose-phosphate
isomerase in infected BALB/C mice. Southeast Asian J Trop Med Publ Health
2004;35:518–22.
[27] Probst P, Stromberg E, Ghalib HW, Mozel M, Badaro R, Reed SG, et al.
Identification and characterization of T cell-stimulating antigens from
Leishmania by CD4 T cell expression cloning. J Immunol 2001;166:498–505.
[28] Garg R, Srivastava JK, Pal A, Naik S, Dube A. Isolation of integral membrane
proteins of Leishmania promastigotes and evaluation of their prophylactic
potential in hamsters against experimental visceral leishmaniasis. Vaccine.
2005;23:1189–96.
[29] Dube A, Singh N, Sundar S, Singh N. Refractoriness to the treatment of sodium
stibogluconate in Indian kala-azar field isolates persist in in vitro and in vivo
experimental models. Parasitol Res 2005;96:216–23.
[30] Gupta SK, Sisodia BS, Sinha S, Hajela K, Naik S, Shasany AK, et al. Proteomic
approach for identification and characterization of novel immunostimulatory
proteins from soluble antigens of Leishmania donovani promastigotes.
Proteomics 2007;7:816–23.
[31] Bradford MM. A rapid and sensitive method for the quantitation of microgram
quantities of protein utilizing the principle of protein-dye binding. Anal
Biochem 1976;72:248–54.
[32] Mitra S, Ghosh L, Chakrabarty P, Biswas M, Bhattacharyya FK, Ghosh DK. Effect
of bioamines on uptake of promastigotes of Leishmania donovani by hamster
peritoneal macrophages. J Med Microbiol 1992;36:283–7.
[33] Morgan DM. Tetrazolium (MTT) assay for cellular viability and activity.
Methods Mol Biol 1998;79:179–83.
[34] Zivcec M, Safronetz D, Haddock E, Feldmann H, Ebihara H. Validation of assays
to monitor immune responses in the Syrian golden hamster (Mesocricetus
auratus). J Immunol Methods 2011;368:24–35.
[35] Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-
time quantitative PCR and the 2("Delta Delta C(T)) method. Methods (San
Diego, Calif) 2001;25:402–408.
[36] Jaffe CL, Rachamim N. Amastigote stage-specific monoclonal antibodies
against Leishmania major. Infect Immun 1989;57:3770–7.
2298 Keerti et al. / Vaccine 36 (2018) 2293–2299
[37] Mullen AB, Baillie AJ, Carter KC. Visceral leishmaniasis in the BALB/c mouse: a
comparison of the efficacy of a nonionic surfactant formulation of sodium
stibogluconate with those of three proprietary formulations of amphotericin B.
Antimicrob Agents Chemother. 1998;42:2722–5.
[38] Carter KC, Baillie AJ, Alexander J, Dolan TF. The therapeutic effect of sodium
stibogluconate in BALB/c mice infected with Leishmania donovani is organ-
dependent. J Pharmacy Pharmacol 1988;40:370–3.
[39] Rasouli M, Karimi MH, Kalani M, Ebrahimnezhad S, Namayandeh M, Moravej
A. Immunostimulatory effects of Leishmania infantum HSP70 recombinant
protein on dendritic cells in vitro and in vivo. Immunotherapy 2014;6:577–85.
[40] Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via arginase or nitric
oxide synthase: two competing arginine pathways in macrophages. Front
Immunol 2014:5.
[41] Kamhawi S, Belkaid Y, Modi G, Rowton E, Sacks D. Protection against
cutaneous leishmaniasis resulting from bites of uninfected sand flies.
Science (New York, NY) 2000;290:1351–4.
[42] Gifawesen C, Farrell JP. Comparison of T-cell responses in self-limiting versus
progressive visceral Leishmania donovani infections in golden hamsters. Infect
Immun 1989;57:3091–6.
[43] Islamuddin M, Chouhan G, Want MY, Ozbak HA, Hemeg HA, Afrin F.
Immunotherapeutic potential of eugenol emulsion in experimental visceral
leishmaniasis. PLoS Negl Trop Dis 2016;10:e0005011.
[44] Liew FY, Li Y, Millott S. Tumor necrosis factor-alpha synergizes with IFN-
gamma in mediating killing of Leishmania major through the induction of
nitric oxide. J Immunol 1990;145:4306–10.
[45] Lopes MF, Costa-da-Silva AC, DosReis GA. Innate Immunity to leishmania
infection: within phagocytes. Mediat Inflamm 2014:2014.
[46] Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to
Leishmania major in mice. Nat Rev Immunol 2002;2:845–58.
[47] Rodrigues V, Cordeiro-da-Silva A, Laforge M, Silvestre R, Estaquier J. Regulation
of immunity during visceral Leishmania infection. Parasit Vectors 2016;9:118.
[48] Gautam S, Kumar R, Maurya R, Nylen S, Ansari N, Rai M, et al. IL-10
neutralization promotes parasite clearance in splenic aspirate cells from
patients with visceral leishmaniasis. J Infect Dis 2011;204:1134–7.
Keerti et al. / Vaccine 36 (2018) 2293–2299 2299
... Aldolase (rLdAld) and Enolase (rLdEno), alone exhibited an enhanced immunotherapeutic effect against established Leishmania infection. 17 These proteins noticeably modulated the immune status of infected animals by stimulating the cellular defensive responses such as an enhanced DTH reaction and skewed Th1 cytokine profile. Further, liposomal amphotericin B (Ambisome), a recommended antileishmanial to treat VL patients, 18 reports to be associated with relapses and demands new short course multiple drug therapies. ...
... 22 Thereafter, purified proteins tested for the endotoxin content using Limulus amoebocyte lysate test kit (Thermo Fisher, USA) and concentrated to a smaller volume following prior protocols. 17 Soluble Leishmania antigen (SLD) was also extracted from the log phase culture of promastigotes as per the protocol of Gupta et al., 2007 and kept at À80 C until further use. 23 Immunization schedule 50 female hamsters were divided into five groups of ten hamsters in each group. ...
... DTH response was assessed as footpad thickness in mm units by measuring the difference in footpad swelling between the two footpads (with and without SLD) in each hamster. 17 ...
Article
Background Available therapeutics for visceral leishmaniasis (VL), a deadly parasitic infection, are usually associated with inadequate efficacy and adverse aftereffects. Further, the primary site of Leishmania parasite are host macrophages resulting in compromised immunity; ensuing marked T-cell immunosuppression. Such settings emphasize the exploration of chemo-immunotherapeutic strategies for improvising the infected person's immune status with better resolution of infection. Methods Present work employs the immunization of Leishmania-infected hamsters with Leishmania-derived recombinant aldolase (rLdAld) and enolase (rLdEno) proteins in consort with the sub-optimal dose of Ambisome (2.5 mg/kg). After the completion of immunization, hamsters were sacrificed on day 60 and 90 post infection and different organ samples were collected to perform immunological assay for evaluating the therapeutic efficacy and modulation in protective cellular immune responses. Results Combining these proteins, particularly rLdAld with Ambisome (2.5 mg/kg), has significantly reduced the parasitic load (∼80%) with remarkable enhancement in DTH and lymphoproliferative responses compared to the infected control and only Ambisome treated groups. Moreover, cytokine levels at RNA and protein levels were noticed to be inclined towards Th-1 phenotype through up-regulation of IFN-γ and TNF-α with significant down-regulation in IL-10 and TGF-β expression, an indication towards the generation of protective immunity against experimental VL. Conclusion Our experimental findings demonstrated that the chemo-immunotherapeutic approach could be an effective way of controlling human VL infection at minimal dosages of antileishmanial with reduced side-effects and propensity of drug resistance emergence.
... parasite burden in treated animals (Da Silva et al. 2020a, b). Several immunotherapeutic regimens by the use of Leishmania antigens either alone or in association with conventional therapy have been tested, showing varying degrees of efficacy, as well as the reduction of dog infectivity to the vector (Guarga et al. 2002;Borja-Cabrera et al. 2004;Saraiva et al. 2006;Santos et al. 2007;Trigo et al. 2010;Jamshidi et al. 2011;Roatt et al. 2017;Keerti et al. 2018;Toepp et al. 2018;Gonçalves et al. 2019). However, the association of either allopurinol or miltefosine with rLdccys1 did not lead to a significant difference in parasite reduction compared to that resulting from the treatment with these drugs alone. ...
Article
Full-text available
The purpose of the present study was to evaluate the efficacy of the treatment with a recombinant cysteine proteinase from Leishmania, rldccys1, associated with allopurinol or miltefosine on Leishmania (Leishmania) infantum chagasi–infected hamsters. Golden Syrian hamsters infected with L. (L.) infantum chagasi were treated with either miltefosine (46 mg/kg) or allopurinol (460 mg/kg) alone by oral route or associated with rldccys1 (150 µg/hamster) by subcutaneous route for 30 days. Infected hamsters were also treated with miltefosine (46 mg/kg) plus rldccys1 (150 µg/hamster) for 30 days (phase 1) followed by two additional doses of rldccys1 (250 µg/hamster) (phase 2). After the end of treatment, the animals were analyzed for parasite load, body weight, serum levels of immunoglobulins, cytokine expression, and drug toxicity. The data showed a significant decrease of parasite load in infected hamsters treated with allopurinol or miltefosine alone or associated with rldccys1, as well as in those treated with rldccys1 alone. Significantly lower levels of serum IgG were detected in hamsters treated with allopurinol plus rldccys1. The treatment with miltefosine associated with rldccys1 prevented relapse observed in animals treated with miltefosine alone. A significant loss of body weight was detected only in some hamsters treated with miltefosine for 1 month and deprived of this treatment for 15 days. There were no significant differences in transcript expression of IFN-γ and IL-10 in any of treated groups. Neither hepatotoxicity nor nephrotoxicity was observed among controls and treated groups. These findings open perspectives to further explore this immunochemotherapeutic schedule as an alternative for treatment of visceral leishmaniasis.
... Understanding the basis of protective immune responses to Leishmania infection guides the research and development of candidate vaccines. Several vaccine candidates against VL have been evaluated mainly in murine and/or canine models [8][9][10][11][12][13]. They include dead parasites administered with or without adjuvants, genetically modified parasites, or viruses expressing Leishmania genes encoding for recombinant proteins, plasmid DNA-based vaccines that encode genes in eukaryotic expression [14,15], and protein-based vaccines using whole recombinant proteins and chimera proteins containing T cell epitopes [16][17][18][19]. ...
Chapter
Leishmaniases are neglected diseases caused by Leishmania parasites and affect millions of people worldwide. The induction of protective immunity against infection by some species of Leishmania has stimulated the development of vaccine candidates against the disease. In this chapter we describe protocols for immunizing mice with a recombinant chimera vaccine containing selected epitopes that specifically stimulate a Th1-type immune response. We describe protocols for challenging mice with live Leishmania parasite and for measuring parameters of the immune response to vaccination and parasite infection, including the production of cytokines, nitric oxide, and IgG antibodies, and the contribution of CD4+ and CD8+ T cells. We also provide protocols for isolating mouse organs for cell culture and for quantifying parasite loads in unvaccinated control animals and in vaccine-protected animals. These protocols can form the basis of immunological studies of candidate Leishmania vaccines in the mouse, as a step toward further vaccine development for human use.
... It does corroborate with a study in which a treatment consisting of an immuno-modulator (liposomal CpG- ODN-2006) and miltefosine in L. donovani-infected hamsters showed the dominance of Th1-type immune response with an increase of inducible nitric oxide synthase (Shivahare et al., 2014). Another study showed that hamsters immunotreated with recombinant proteins of L. donovani presented an increase of IFN-γ and TNF-α cytokines expression (Rawat et al., 2018). Similar results, as higher levels of pro-inflammatory cytokines and decrease of interleukin-10 in spleen, also were demonstrated in hamsters treated with Miltefosine or its derivative (Gupta et al., 2012;da Silva et al., 2020). ...
Article
Full-text available
The control of human visceral leishmaniasis (VL) is hard since there are no vaccines available as well as the treatment is hampered by toxicity and resistant parasites. Furthermore, as human, and canine VL causes immunosuppression, the combination of drugs with immunostimulatory agents is interesting to upregulate the immunity, reducing side-effects, improving treatment approaches against disease. Herein, we assessed the immunochemotherapy using miltefosine along with a vaccine formulated by Leishmania braziliensis antigens + saponin + monophosphoryl lipid-A (LBSapMPL) in L. infantum -infected hamsters. Two months after infection, the animals received treatments, and after 15 days they were evaluated for the treatment effect. The potential anti- Leishmania effect of miltefosine + LBSapMPL-vaccine was revealed by a specific immune response activation reflecting in control of spleen parasitism using half the miltefosine treatment time. The treated animals also showed an increase of total and T-CD4 splenocytes producing IFN- γ and TNF- α and a decrease of interleukin-10 and anti-Leishmania circulating IgG. In addition, it was demonstrated that the control of spleen parasitism is related to the generation of a protective Th1 immune response. Hence, due to the combinatorial action of miltefosine with LBSapMPL-vaccine in immunostimulating and controlling parasitism, this immunochemotherapy protocol can be an important alternative option against canine and human VL.
... Cases of VL mainly occur in South Asia, East Africa (South Sudan, Sudan, Somalia), Iraq, Brazil, and Ethiopia. The majority (about 90%) of cases occur in five countries: Sudan, Nepal, India, Brazil, and Ethiopia (Keerti et al., 2018). ...
Article
Coinfection is less commonly observed in individuals around the world, yet it is more common than the single infection. Around 800 million people worldwide are infected with helminths as a result of various diseases. Lymphatic filariasis (LF) and visceral leishmaniasis (VL) are chronic, deadly, crippling, and debilitating neglected tropical diseases (NTDs) that are endemic in tropical and subtropical regions of the world. Due to poor hygienic conditions, poverty, and genetic predisposition, those living in endemic areas are more likely to develop both leishmaniasis and filariasis. One of the key challenges in the management of LF/VL coinfection is the development of an effective therapeutic strategy that not only treats the first episode of VL but also prevents LF. However, there is a scarcity of knowledge and data on the relationship between LF and VL coinfection. While reviewing it was apparent that only a few studies relevant to LF/VL coinfections have been reported from southeastern Spain, Sudan, and the Indian subcontinents, highlighting the need for greater research in the most affected areas. We also looked at LF and VL as a single disease and also as a coinfection. Some features of the immune response evolved in mammalian hosts against LF and VL alone or against coinfection are also discussed, including epidemiology, therapeutic regimens, and vaccines. In addition to being potentially useful in clinical research, our findings imply the need for improved diagnostic methodology and therapeutics, which could accelerate the deployment of more specific and effective diagnosis for treatments to lessen the impact of VL/LF coinfections in the population.
Chapter
Visceral leishmaniasis (VL), a neglected tropical disease, is caused by the parasite Leishmania donovani complex. Whereas the transmission of L. donovani infection is anthroponotic in the Indian subcontinent and East Africa, the spread of Leishmania infantum, as well as Leishmania chagasi, is zoonotic, with dogs serving as the reservoir host throughout Europe, North Africa, and regions of Latin America. Although zoonotic VL is becoming less common, anthroponotic VL continues to cause epidemics periodically. These parasites infect the host’s macrophages and damage the immune system, leading to pronounced immunosuppression in the affected person. In general, a small percentage of L. donovani-infected population develop full-blown clinical disease and are treated, but the majority of them remain asymptomatic. In addition, the treated VL patients develop post-kala-azar dermal leishmaniasis (PKDL) conditions, and these populations contribute to spreading the VL disease via the sandfly vector. This condition is one of the main obstacles to the World Health Organization’s efforts to eradicate this fatal disease. Designing therapeutic vaccines that can only target infected macrophages has been attempted in the absence of any reliable and efficient vector control measures. Three vaccines—Leishmune®, Leish-Tec®, and Leish-111f® with monophosphoryl lipid A (MPLA) plus squalene emulsion in combination with glucantime—have thus far effectively undergone immunotherapeutic evaluation against canine VL (CVL). A third-generation vaccine—ChAd63 KH has also shown its immunotherapeutic potential against PKDL. Nevertheless, it is still necessary to translate positive findings from a number of experimental studies against human VL into therapeutic applications. This chapter summarizes the numerous approaches that have been attempted and evaluated for generating therapeutic vaccines to stave off this dreaded disease.
Article
Full-text available
Control of the intracellular parasite Leishmania (L.) requires the activation of strong type 1 cellular immune responses. Towards this goal, in the present study, a multiepitope chimeric protein named LiChimera was encapsulated into cationic liposomes and its protective efficacy against experimental visceral leishmaniasis was investigated. Liposomal LiChimera conferred significant protection against L. infantum as evidenced by the significantly reduced parasite loads in the spleen and liver. Protection detected in Lipo:LiChimera-immunized mice was dependent on the differentiation of long-lasting cellular immune responses and particularly the induction of antigen-specific multifunctional memory CD4+ TH1 and CD8+ T cells that persisted during infection, as evidenced by the persistent high production of IFN-γ and IL-2 and proliferation activity. Notably, protected mice were also characterized by significantly low numbers of non-regulatory CD4+ T cells able to co-produce IFN-γ and IL-10, an important population for disease establishment, as compared to non-immunized control group. Collectively, these results demonstrate that cationic liposomes containing LiChimera can be considered an effective candidate vaccine against visceral leishmaniasis.
Article
Leishmaniasis is an enigmatic disease that has very restricted options for chemotherapy and none for prophylaxis. As a result, deriving therapeutic principles for curing the disease has been a major objective in Leishmania research for a long time. Leishmania is a protozoan parasite that lives within macrophages by subverting or switching cell signaling to the pathways that ensure its intracellular survival. Therefore, three groups of molecules aimed at blocking or eliminating the parasite, at least, in principle, include blockers of macrophage receptor- Leishmania ligand interaction, macrophage-activating small molecules, peptides and cytokines, and signaling inhibitors or activators. Macrophages also act as an antigen-presenting cell, presenting antigen to the antigen-specific T cells to induce activation and differentiation of the effector T cell subsets that either execute or suppress anti-leishmanial functions. Three groups of therapeutic principles targeting this sphere of Leishmania-macrophage interaction include antibodies that block pro-leishmanial response of T cells, ligands that activate anti-leishmanial T cells and the antigens for therapeutic vaccines. Besides these, prophylactic vaccines have been in clinical trials but none has succeeded so far. Herein, we have attempted to encompass all these principles and compose a comprehensive review to analyze the feasibility and adoptability of different therapeutics for leishmaniasis.
Article
Haemaphysalis longicornis is an obligate hematophagous ectoparasite that transmits a variety of pathogens causing life‑threatening diseases in humans and animals. Triosephosphate isomerase (TIM) is a key enzyme in glycometabolism, making in an interesting anti-tick vaccine candidate antigen. In this study, the open reading frame (ORF) of the TIM homologue from H. longicornis (HlTIM) was shown to consist of 747 bp encoding a protein of 248 amino acids. HlTIM gene expression was detected in all developmental stages and in all tissues of the unfed female tick by quantitative real-time PCR. The HlTIM gene was cloned and inserted into pET-32a (+) to obtain the recombinant HlTIM protein (rHlTIM) and its immunogenicity was confirmed by Western blot. ELISA results showed that rabbits immunized with rHlTIM produced a humoral immune response. A vaccine trial in rabbits against H. longicornis infestation demonstrated that the engorgement weight, oviposition and hatchability of ticks from the rH1TIM group was decreased by 8.6%, 35.4% and 17.3% respectively, compared to the histidine-tagged thioredoxin (Trx) control group. Considering the cumulative effect of vaccination on the evaluated parameters, results showed 50.9% efficacy in the rHlTIM group. The data reported here demonstrate that rHlTIM has potential for further development of a new candidate vaccine for tick control.
Article
Full-text available
Herein, we evaluated the treatment strategy employing a therapeutic heterologous vaccine composed of antigens of Leishmania braziliensis associated with MPL adjuvant (LBMPL vaccine) for visceral leishmaniasis (VL) in symptomatic dogs naturally infected by Leishmania infantum. Sixteen dogs received immunotherapy with MPL adjuvant (n = 6) or with a vaccine composed of antigens of L. braziliensis associated with MPL (LBMPL vaccine therapy, n = 10). Dogs were submitted to an immunotherapeutic scheme consisting of 3 series composed of 10 subcutaneous doses with 10-day interval between each series. The animals were evaluated before (T0) and 90 days after treatment (T90) for their biochemical/hematological, immunological, clinical, and parasitological variables. Our major results showed that the vaccine therapy with LBMPL was able to restore and normalize main biochemical (urea, AST, ALP, and bilirubin) and hematological (erythrocytes, hemoglobin, hematocrit, and platelets) parameters. In addition, in an ex vivo analysis using flow cytometry, dogs treated with LBMPL vaccine showed increased CD3⁺ T lymphocytes and their subpopulations (TCD4⁺ and TCD8⁺), reduction of CD21⁺ B lymphocytes, increased NK cells (CD5⁻CD16⁺) and CD14⁺ monocytes. Under in vitro conditions, the animals developed a strong antigen-specific lymphoproliferation mainly by TCD4⁺ and TCD8⁺ cells; increasing in both TCD4⁺IFN-γ⁺ and TCD8⁺IFN-γ⁺ as well as reduction of TCD4⁺IL-4⁺ and TCD8⁺IL-4⁺ lymphocytes with an increased production of TNF-α and reduced levels of IL-10. Concerning the clinical signs of canine visceral leishmaniasis, the animals showed an important reduction in the number and intensity of the disease signs; increase body weight as well as reduction of splenomegaly. In addition, the LBMPL immunotherapy also promoted a reduction in parasite burden assessed by real-time PCR. In the bone marrow, we observed seven times less parasites in LBMPL animals compared with MPL group. The skin tissue showed a reduction in parasite burden in LBMPL dogs 127.5 times higher than MPL. As expected, with skin parasite reduction promoted by immunotherapy, we observed a blocking transmission to sand flies in LBMPL dogs with only three positive dogs after xenodiagnosis. The results obtained in this study highlighted the strong potential for the use of this heterologous vaccine therapy as an important strategy for VL treatment.
Article
Full-text available
Background The therapy of visceral leishmaniasis (VL) is limited by resistance, toxicity and decreased bioavailability of the existing drugs coupled with dramatic increase in HIV-co-infection, non-availability of vaccines and down regulation of cell-mediated immunity (CMI). Thus, we envisaged combating the problem with plant-derived antileishmanial drug that could concomitantly mitigate the immune suppression of the infected hosts. Several plant-derived compounds have been found to exert leishmanicidal activity via immunomodulation. In this direction, we investigated the antileishmanial activity of eugenol emulsion (EE), complemented with its immunomodulatory and therapeutic efficacy in murine model of VL. Methodology/Principal Findings Oil-in-water emulsion of eugenol (EE) was prepared and size measured by dynamic light scattering (DLS). EE exhibited significant leishmanicidal activity with 50% inhibitory concentration of 8.43±0.96 μg ml⁻¹ and 5.05±1.72 μg ml─1, respectively against the promastigotes and intracellular amastigotes of Leishmania donovani. For in vivo effectiveness, EE was administered intraperitoneally (25, 50 and 75 mg/kg b.w./day for 10 days) to 8 week-infected BALB/c mice. The cytotoxicity of EE was assessed in RAW 264.7 macrophages as well as in naive mice. EE induced a significant drop in hepatic and splenic parasite burdens as well as diminution in spleen and liver weights 10 days post-treatment, with augmentation of 24h-delayed type hypersensitivity (DTH) response and high IgG2a:IgG1, mirroring induction of CMI. Enhanced IFN-γ and IL-2 levels, with fall in disease-associated Th2 cytokines (IL-4 and IL-10) detected by flow cytometric bead-based array, substantiated the Th1 immune signature. Lymphoproliferation and nitric oxide release were significantly elevated upon antigen revoke in vitro. The immune-stimulatory activity of EE was further corroborated by expansion of IFN-γ producing CD4⁺ and CD8⁺ splenic T lymphocytes and up-regulation of CD80 and CD86 on peritoneal macrophages. EE treated groups exhibited induction of CD8⁺ central memory T cells as evidenced from CD62L and CD44 expression. No biochemical alterations in hepatic and renal enzymes were observed. Conclusions Our results demonstrate antileishmanial activity of EE, potentiated by Th1 immunostimulation without adverse side effects. The Th1 immune polarizing effect may help to alleviate the depressed CMI and hence complement the leishmanicidal activity.
Article
Full-text available
A major problem with tuberculosis (TB) control is the long duration of drug therapy?both for latent and for active TB. Therapeutic vaccination has been postulated to improve this situation, and to this end there are several candidates already in clinical phases of development. These candidates follow two main designs, namely bacilli-directed therapy based on inactivated -whole or -fragmented bacillus (Mycobacterium w and RUTI) or fusion proteins that integrate non-replicating bacilli -related antigens (H56 vaccine), and host-directed therapy to reduce the tissue destruction. The administration of inactivated Mycobacterium vaccae prevents the ?Koch phenomenon? response, and oral administration of heat-killed Mycobacterium manresensis prevents excessive neutrophilic infiltration of the lesions. This review also tries to explain the success of Mycobacterium tuberculosis by reviewing its evolution from infection to disease, and highlights the lack of a definitive understanding of the natural history of TB pathology and the need to improve our knowledge on TB immunology and pathogenesis.
Article
Full-text available
Our prior studies demonstrated that cellular response of T helper 1 (Th1) type was generated by a soluble antigenic fraction (ranging from 89.9 to 97.1 kDa) of Leishmania donovani promastigote, in treated Leishmania patients as well as hamsters and showed significant prophylactic potential against experimental visceral leishmaniasis (VL). Eighteen Th1 stimulatory proteins were identified through proteomic analysis of this subfraction, out of which 15 were developed as recombinant proteins. In the present work, we have evaluated these 15 recombinant proteins simultaneously for their comparative cellular responses in treated Leishmania patients and hamsters. Six proteins viz. elongation factor-2, enolase, aldolase, triose phosphate isomerase, protein disulfide isomerase, and p45 emerged as most immunogenic as they produced a significant lymphoproliferative response, nitric oxide generation and Th1 cytokine response in PBMCs and lymphocytes of treated Leishmania patients and hamsters respectively. The results suggested that these proteins may be exploited for developing a successful poly-protein and/or poly-epitope vaccine against VL.
Article
Full-text available
Unicellular eukaryotes of the genus Leishmania are collectively responsible for a heterogeneous group of diseases known as leishmaniasis. The visceral form of leishmaniasis, caused by L. donovani or L. infantum, is a devastating condition, claiming 20,000 to 40,000 lives annually, with particular incidence in some of the poorest regions of the world. Immunity to Leishmania depends on the development of protective type I immune responses capable of activating infected phagocytes to kill intracellular amastigotes. However, despite the induction of protective responses, disease progresses due to a multitude of factors that impede an optimal response. These include the action of suppressive cytokines, exhaustion of specific T cells, loss of lymphoid tissue architecture and a defective humoral response. We will review how these responses are orchestrated during the course of infection, including both early and chronic stages, focusing on the spleen and the liver, which are the main target organs of visceral Leishmania in the host. A comprehensive understanding of the immune events that occur during visceral Leishmania infection is crucial for the implementation of immunotherapeutic approaches that complement the current anti-Leishmania chemotherapy and the development of effective vaccines to prevent disease.
Article
Full-text available
The present report documents a 49-year-old HIV-infected man receiving antiretroviral therapy with a suboptimal immune response and a CD4 count of 95 cells/mm 3 , despite virological suppression. Investigation of bone marrow was conducted and yielded a diagnosis of visceral leishmaniasis. The clinical course was complicated by gastrointestinal involvment and relapse occurred after amphotericin B therapy. With the addition of miltefosine, the patient no longer presented with bone marrow amastigotes, and displayed an increased CD4 count and negative Leishmania polymerase chain reaction results. The present case highlights atypical presentation of visceral leishmaniasis, including poor immune reconstitution and gastrointestinal involvement. The high likelihood of relapse and response to combination therapy are illustrated.
Article
Full-text available
Background: Recent studies have shown significant decline in the final cure rate after miltefosine treatment in visceral leishmaniasis. This study evaluates the efficacy of miltefosine in the treatment of post kala-azar dermal leishmaniasis (PKDL) patients recruited over a period of 5 years with 18 months of follow-up. Methodology: In this study 86 confirmed cases of PKDL were treated with two different dosage regimens of miltefosine (Regimen I- 50mg twice daily for 90 days and Regimen II- 50 mg thrice for 60 days) and the clinical outcome assessed monthly. Cure/relapse was ascertained by clinical and histopathological examination, and measuring parasite burden by quantitative real-time PCR. In vitro susceptibility of parasites towards miltefosine was estimated at both promastigote and amastigote stages. Results: Seventy three of eighty six patients completed the treatment and achieved clinical cure. Approximately 4% (3/73) patients relapsed by the end of 12 months follow-up, while a total of 15% (11/73) relapsed by the end of 18 months. Relapse rate was significantly higher in regimen II (31%) compared to regimen I (10.5%)(P<0.005). Parasite load at the pre-treatment stage was significantly higher (P<0.005) in cases that relapsed compared to the cases that remained cured. In vitro susceptibility towards miltefosine of parasites isolated after relapse was significantly lower (>2 fold) in comparison with the pre-treatment isolates (P<0.005). Conclusion: Relapse rate in PKDL following miltefosine treatment has increased substantially, indicating the need of introducing alternate drugs/ combination therapy with miltefosine.
Article
Full-text available
Leishmaniasis is a parasitic disease that encompasses a range of clinical manifestations affecting people in tropical and subtropical regions of the world. Epidemiological and experimental data indicate that protection from disease can be achieved in most people. In addition, we know how the host immune system must respond to infection in order to control parasite growth. However, there is still no vaccine for use in humans. Here, we review our understanding of host immunity following Leishmania infection and also discuss recent advances in the development of vaccines to prevent leishmaniasis, highlighting a new promising approach that targets the parasite hemoglobin receptor.
Article
A number of leishmaniasis vaccine candidates are at various stages of pre-clinical and clinical development. Leishmaniasis is a vector-borne neglected tropical disease (NTD) caused by a protozoan parasite of the genus Leishmania and transmitted to humans by the bite of a sand fly. Visceral leishmaniasis (VL, kala-azar) is a high mortality NTD found mostly in South Asia and East Africa, while cutaneous leishmaniasis (CL) is a disfiguring NTD highly endemic in the Middle East, Central Asia, North Africa, and the Americas. Estimates attribute 50,000 annual deaths and 3.3 million disability-adjusted life years to leishmaniasis. There are only a few approved drug treatments, no prophylactic drug and no vaccine. Ideally, an effective vaccine against leishmaniasis will elicit long-lasting immunity and protect broadly against VL and CL. Vaccines such as Leish-F1, F2 and F3, developed at IDRI and designed based on selected Leishmania antigen epitopes, have been in clinical trials. Other groups, including the Sabin Vaccine Institute in collaboration with the National Institutes of Health are investigating recombinant Leishmania antigens in combination with selected sand fly salivary gland antigens in order to augment host immunity. To date, both VL and CL vaccines have been shown to be cost-effective in economic modeling studies.
Article
Chagas disease, caused by Trypanosoma cruzi, results in an acute febrile illness that progresses to chronic chagasic cardiomyopathy in 30% of patients. Current treatments have significant side effects and poor efficacy during the chronic phase; therefore, there is an urgent need for new treatment modalities. A robust TH1-mediated immune response correlates with favorable clinical outcomes. A therapeutic vaccine administered to infected individuals could bolster the immune response, thereby slowing or stopping the progression of chagasic cardiomyopathy. Prior work in mice has identified an efficacious T. cruzi DNA vaccine encoding Tc24. To elicit a similar protective cell-mediated immune response to a Tc24 recombinant protein, we utilized a poly(lactic-co-glycolic acid) nanoparticle delivery system in conjunction with CpG motif-containing oligodeoxynucleotides as an immunomodulatory adjuvant. In a BALB/c mouse model, the vaccine produced a TH1-biased immune response, as demonstrated by a significant increase in antigen-specific IFNγ-producing splenocytes, IgG2a titers, and proliferative capacity of CD8(+) T cells. When tested for therapeutic efficacy, significantly reduced systemic parasitemia was seen during peak parasitemia. Additionally, there was a significant reduction in cardiac parasite burden and inflammatory cell infiltrate. This is the first study demonstrating immunogenicity and efficacy of a therapeutic Chagas vaccine using a nanoparticle delivery system.