Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Biomedicine & Pharmacotherapy 141 (2021) 111920

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Current status of nanoscale drug delivery and the future of nano-vaccine


development for leishmaniasis – A review
Pragya Prasanna a, 1, Prakash Kumar a, 1, Saurabh Kumar a, Vinod Kumar Rajana a, Vishnu Kant a,
Surendra Rajit Prasad a, Utpal Mohan b, V. Ravichandiran a, b, Debabrata Mandal a, *
a
Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, India
b
National Institute of Pharmaceutical Education and Research, Kolkata 700054, India

A B S T R A C T

The study of tropical diseases like leishmaniasis, a parasitic disease, has not received much attention even though it is the second-largest infectious disease after
malaria. As per the WHO report, a total of 0.7–1.0 million new leishmaniasis cases, which are spread by 23 Leishmania species in more than 98 countries, are
estimated with an alarming 26,000–65,000 death toll every year. Lack of potential vaccines along with the cost and toxicity of amphotericin B (AmB), the most
common drug for the treatment of leishmaniasis, has raised the interest significantly for new formulations and drug delivery systems including nanoparticle-based
delivery as anti-leishmanial agents. The size, shape, and high surface area to volume ratio of different NPs make them ideal for many biological applications. The
delivery of drugs through liposome, polymeric, and solid-lipid NPs provides the advantage of high biocomatibilty of the carrier with reduced toxicity. Importantly,
NP-based delivery has shown improved efficacy due to targeted delivery of the payload and synergistic action of NP and payload on the target. This review analyses
the advantage of NP-based delivery over standard chemotherapy and natural product-based delivery system. The role of different physicochemical properties of a
nanoscale delivery system is discussed. Further, different ways of nanoformulation delivery ranging from liposome, niosomes, polymeric, metallic, solid-lipid NPs
were updated along with the possible mechanisms of action against the parasite. The status of current nano-vaccines and the future potential of NP-based vaccine are
elaborated here.

1. Introduction have studied the cytotoxicity of NPs by studying NP- protein corona
level (protein adsorption layers located on the surface of colloidal NPs
Nanotechnology has gained importance in a wide range of applica­ level [5]. Advances in nanotechnology have provided numerous drug
tions because it can be tuned to attain desirable attributes by manipu­ delivery options to reduce dosage, enable improved release promoting
lating physicochemical properties like size, shape, charge, etc. [1]. The high drug concentrations at the target sites. For instance, specific and
NPs are considered as the fundamental element of nanotechnology targeted macrophage delivery will be highly beneficial in case of
whose size varies in the wide range of 1–1000 nm and beyond with intracellular infections like leishmaniasis [6,7].
multiple applications in biology and medicine [2]. Nowadays a trend of Leishmaniasis is a spectrum of diseases primarily prevalent in poor
synthesizing NPs using biomaterials such as plants and microorganisms and underdeveloped economies with an estimated 7,00,000–1 million
is regarded as a reliable approach where both gold and silver NPs of cases reported annually (WHO) [8]. The causative agent of leishmani­
varied shapes and sizes have been synthesised using plants extracts. asis, Leishmania is transmitted by the bite of infected female phleboto­
These NPs are advantageous in terms of greater antimicrobial activity mine sandflies and it completes its life cycle in two different hosts: sand
and reduced toxicity compared to chemically synthesized NPs [3]. flies and vertebrates [9]. Therapeutic agents like antimonials, Ampho­
Further, antimicrobial textiles have also been developed by attaching tericin B (AmB), paromomycin, sitamaquine, and miltefosine became
NPs in the fibers to make self-decontaminating antimicrobial textiles insufficient due to side effects like toxicity, increased drug resistance,
[4]. For the safety and toxicity issues, the study of NPs- protein in­ and in some cases due to cost-associated with its formulation. The heavy
teractions are also required. There are multiple proteomic studies that metal, antimony, in the pentavalent form (SbV) as sodium antimony

* Correspondence to: Department of Biotechnology, NIPER-Hajipur, Export Promotions Industrial Park (EPIP)-Hajipur, District: Vaishali, Pin 844102 Bihar, India.
E-mail addresses: pragyaprasanna36@gmail.com (P. Prasanna), bbhaskar.kumar73@gmail.com (P. Kumar), sahilsaurabh25@gmail.com (S. Kumar),
vinoddhniper@gmail.com (V.K. Rajana), vishnukant378@gmail.com (V. Kant), surendra.science@gmail.com (S.R. Prasad), mohan.utpal@gmail.com (U. Mohan),
directorniperkolkata@gmail.com (V. Ravichandiran), debabrataman@gmail.com (D. Mandal).
1
Contributed equally in this manuscript.

https://doi.org/10.1016/j.biopha.2021.111920
Received 5 May 2021; Received in revised form 9 July 2021; Accepted 12 July 2021
Available online 20 July 2021
0753-3322/© 2021 Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

gluconate (SAG) has been the basis of drug treatment for visceral facilitates the improved and sustained release of cargo at the specific
leishmaniasis (VL) since the 1940s. However, clinical resistance to SAG organ or tissues where it is destined. Encapsulation of drugs into
has been a problem in India since the late 1990s and a cure rate of 60% nanocarriers, also, provides enhanced protection against intracellular
was reported [10]. Later, the region endemic for VL in North Bihar, and extracellular degradation compared to naked drugs [27].
India, got the unique distinction of being the only region in the world Nanoformulation-based targeted drug delivery has shown promise to
where widespread primary failure to SAG has been reported [11] overcome drug resistance associated with leishmaniasis [28]. Multiple
causing complete discontinuation of SAG for the treatment of VL. drug delivery strategies targeting drug resistance strains have been
AmBisome, a liposomal formulation of AmB, is the most widely accepted formulated with considerable success in vitro and in vivo using animal
treatment to date for the disease VL which is caused by the parasite models [23]. Different liposomes and hybrid NPs loaded with anti­
Leishmania donovani [12] although it is come with a huge financial leishmanial agents have shown promising results against drug-resistant
burden due to costly lipid-based ingredients [13]. The safety and effi­ strains of the parasite. In most cases, these drug delivery systems
cacy of miltefosine, the only orally administered drug for VL, is also generally target the drug efflux pumps as the emergence of drug resis­
compromised due to the high cost of production and teratogenicity [14]. tance is very often associated with over-expression of these pumps e.g.,
Besides, no registered vaccines are available to date [15]. The challenges ABC transporter, P-glycoprotein containing efflux pumps like MDR1 and
such as limited therapeutic options, suboptimal diagnostics, poor com­ MRP1 [28]. High drug efflux causes a decrease in the intracellular
munity awareness, drug resistance, toxicity, asymptomatic carriers, and, concentrations of the internalized drug subsequently decreasing the
most importantly, absence of vaccines encourage more investigation therapeutic effect. Simultaneously drug efflux causes localized toxicity
including nanoscale drug delivery for more effective treatment of of deposited drugs to the healthy tissues which can be evaded using
leishmaniasis. Most important step towards effective treatment of nanoformulation-based delivery [19]. Another beneficial aspect of
leishmaniasis, at present, includes integrating national control strategies nanoscale delivery is the pH adaptability of the drug moieties in mi­
with simple, reliable, and affordable field serological test along with the croenvironments. The majority of antileishmanial agents are adminis­
creation of cost-effective and affordable diagnostic kits, drugs or vac­ tered through parenteral mode. The paucity of orally administered
cines considering the socio-economic problem associated with the dis­ antileishmanial demands alternatives with high aqueous solubility and
ease [16,17]. stability at gastrointestinal pH. There are several lipid-based delivery
The application of nanoformulation using antileishmanial drugs for systems like palmitoyl homocysteine [29,30], hyaluronic hydrogels,
improved delivery started in 1992 [7]. Since then, many NFs both in the stealth polyethylene glycol (PEG) layers, and pH-sensitive linkers which
form of drugs and vaccines have been prepared and tested at preclinical can be used to prepare pH-sensitive delivery vehicles favoring drug
levels for leishmaniasis [18,19]. However, low success rates of nano­ release in a wide pH range [31]. This is important since the parasite
scale delivery system in vivo and at clinical studies so far, encouraged us resides in the macrophages of the liver and spleen of the infected host
to analyze the possible reasons for the failure of NP-based delivery where the pH is low from its bulk counterpart.
system and a possible future for the same.
3. Important physiochemical advantages of nanoscale drug
2. Importance of nanoscale delivery systems delivery

NPs are the most fundamental component in the fabrication of a 3.1. Shape and size of particles
nanostructure, which behaves differently from its bulk counterparts
[20]. Initially, the idea of nano-based drug delivery arose to address the NPs can be synthesized in different shapes and sizes [20]. These
limitations and harmful effects of formulation excipients caused due to variations in turn determine the toxicity and efficacy of NPs. Several
fast and random distribution as well as toxic consequences by acting as studies have highlighted that size, shape, charge, and surface coating of
vectors for drugs that have improved pharmacokinetic parameters e.g. NPs are major determinants of uptake and toxicity that could be
systemic bioavailability. Nowadays, the branches of nanotechnology are manipulated solely to make them non-toxic NPs. Spherical NPs are
being developed especially to overcome biological barriers such as comparatively less toxic than other dimensions whereas platelet-shaped
gastrointestinal pH, blood-brain barrier, etc. [21,22]. NP-based drug AgNPs are detrimental to epithelial cells [32]. Spherical GNPs and both,
delivery systems have advantages such as high stability and loading with spherical and cubic ZnO NPs, had shown lower cytotoxicity than other
the cargo, ease of incorporation of both hydrophilic and hydrophobic structural forms [33]. The size of the NP is important for cellular uptake,
substances, feasibility of variable routes of administration, including its interactions with the immune system, and its clearance from the body
oral and inhalation routes which makes better treatment options for [34]. In VL the parasite primarily infects the macrophages of the liver
many diseases including leishmaniasis [23]. Furthermore, they provide and spleen. Therefore, uptake studies of macrophage cell lines using
the luxury of improvement of drug bioavailability and other pharma­ different NPs could provide a strategy for improved drug delivery for VL
cokinetic parameters conveniently without any loss of drug efficacy. [35]. Using three different types of NPs (stars, rods, and triangles) of the
NP-based drug delivery act as a vehicle to alter the physio-chemical same size it was observed that uptake was much higher for triangular
properties of a drug easily. For instance, to improve the aqueous solu­ NPs than the others in murine macrophages [36]. The clathrin-mediated
bility of poorly water-soluble drugs, a water-soluble polymer or metallic endocytosis pathway was found to be important in the uptake of gold
NP-based delivery system can be easily linked with the drug molecule NPs. The anionic surface coated triangular GNPs were compared with
covalently or non-covalently without any significant loss in its biological spherical GNPs for their uptake in mouse macrophage cell line RAW
activity [24]. Besides, these delivery systems would, most often, also 264.7 and Hela cell lines. It was found that triangular GNPs were
protect drugs from quick inactivation/degradation in circulation. internalized more with increasing size whereas spherical GNPs are
Increasing the aqueous solubility of drug moiety through chemical internalized more with decreasing size [37]. The synthesized GNPs of
modification is a daunting task and has a risk of loss of its bioactivity. sub-10 nm size (2,4 and 6 nm) show internalization based on size and
The same process with the help of nanotechnology-based modifications nature of surface charge [38]. For zwitterionic and anionic GNPs, uptake
of drug molecules is easier and restores the inherent drug properties. decreased with increasing size, whereas for cationic NPs uptake de­
Since the delivery vehicle is a separate entity that carries the drugs as creases with reducing size. In another study, macrophage uptake was
cargo, it can be conjugated with receptors. ligands and signaling mole­ found to be more for PEG-coated gold nanospheres than nanorods [39].
cules destined for a targeted tissue/organ delivery subsequently These studies show the importance of NP geometry and surface prop­
reducing the dosage by promoting increased drug accumulations at the erties on transport across biological barriers. These studies also estab­
target sites [25,26]. Engineering of these drug delivery systems also lished that uptake of NPs in macrophages are not only dependent on size

2
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

but also on the geometry of NPs and associated surface charge. biological barrier. Commonly used surface modifiers of metal NPs are
disulfide, amine, thiolate, dithioline, carboxylate, and phosphine
3.2. Surface charge of NPs groups. Modification by PEG reduces the cytotoxicity whereas attaching
carboxyl groups (-COOH) increase hydrophilic properties thereby
Surface charge is important for uptake and biodistribution of NPs in altering the internalization of drugs [39]. The polymeric NPs and lipo­
vivo [40]. NPs are routinely tested for use in medical products, like in somes interact with the macrophages depending on their surface charge
imaging, gene, and drug delivery. For these applications, cellular uptake [45]. Liposomes displaying a negatively charged surface, exhibit a much
is usually important and is governed by surface characteristics such as higher rate of phagocytosis by macrophages as compared to neutral
hydrophobicity and charge along with size. Cationic NPs cause more vesicles [47]. A schematic presentation was given in Fig. 1 regarding the
damage to plasma-membrane integrity, stronger mitochondrial and role of different physicochemical properties including shape, size, and
lysosomal damage, and a higher number of autophagosomes than surface charge in NP-based delivery.
anionic NPs. It was observed that nonphagocytic cells uptake cationic
NPs to a higher extent, but charge density and hydrophobicity are 3.3. pH-dependent targeting of NPs
equally important. Phagocytic cells have a preference to take up anionic
NPs [41]. Indeed, negatively charged phosphatidylserine (PS) in the Leishmania species after infection live in an acidic phagolysosomal
outer leaflet of the Leishmania spp. the plasma membrane was found to compartment where the pH is low. A drug moiety capable of reaching a
be important for its attachment to macrophages [42,43]. Cells do not use target should resist the variable pH seen across different tissues or
different uptake mechanisms for cationic and anionic NPs, but high subcellular compartments and, therefore, pH-dependent drug release
uptake rates are always associated with increased biological effects. will be very important for the site-specific accumulation and action of
Phagocytic THP-1 cells or non-phagocytic A549 cells when incubated drugs. pH-responsive NP platform with polymer as the inner core,
with fluorophore-conjugated polystyrene NPs (F-PLNPs) show differ­ amphiphilic lipid-poly(ethylene glycol) (lipid-PEG) as the outer shell,
ential uptake which correlated well with zeta potential [44]. This result and a peptide encoded surface for efficient internalization and sharp
shows that surface charge is a critical parameter in determining cellular endosomal pH response has been reported [48]. NPs consisting of a
uptake efficiency, although other factors such as aggregation/agglom­ drug-loaded polylactic-co-glycolic acid (PLGA) core and wrapped with
eration, protein corona formation which, in turn, depend on surface acid-triggered membrane peptide have shown facilitated internalization
charge can also influence the cellular uptake partly or indirectly. Neutral into cells within the acidic microenvironment where intracellular con­
NPs have been found to have a slower opsonization rate than charged ditions degrade the NPs, thereby releasing the chemotherapeutic cargo
NPs [45]. Zwitterionic NPs have been found to have prolonged circu­ [49]. The single-walled carbon nanotubes (SWNTs) can selectively
lation. The charge is important for mucoadhesion or diffusion, cellular deliver tyrosine phosphatase inhibitor 1 (TPI) as a drug intracellularly to
uptake, bioavailability, and toxicity [38]. By changes in surface charge macrophages in a pH-Dependent manner [50]. Here it was shown that at
and functionality, the biocompatibility and uptake efficiency can be pH> 7 the drug is stably bound to SWNTs but under acidic conditions at
changed for NPs [46]. Surface modifications using hydro­ pH < 6.0 the drug is released inside the macrophages. So, this kind of pH
philic/hydrophobic components having different ionizable groups specific delivery can be advantageous for the treatment of VL which are
facilitate drug release mechanisms after penetration through the associated with macrophages of the liver, spleen, lymph nodes, etc.

Fig. 1. It represents different physiochemical properties of NPs. (A) Shape (B) Size & its correlation with different biomaterials in nature and (C) Surface charge
associated with functionalization of NPs.

3
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Except for miltefosine, delivery in leishmaniasis lacks drugs that burden with ~80% efficiency than the active alone [64].
could be orally administered. For a drug to be administered orally and Artemisinin-PLGA NPs displayed increased leishmanicidal activity with
exert its effect, it should be able to survive the pH of the gastrointestinal minimal macrophage toxicity compared to free artemisinin [65].
tract that varies throughout fasted and fed state [51] with a consistent Moreover, PLGA NPs of curcumin, a bioactive compound with reported
acidic environment in the stomach (<2) and alkaline in the small in­ leishmanicidal activity were found to be efficacious in combination with
testine (>6) [52]. When pH-sensitive lipids like palmitoyl homocysteine miltefosine at a subcurative dose with negligible nephrotoxicity [66].
(PHC) were incorporated into liposomes composed of dipalmitoyl, Compound 8-hydroxyquinoline (8-HQN) [67,68] in combination with a
stearoyl, and diheptadecanoyl L- α phosphatidylcholine (DPPC, DSPC, polymeric micelle system showed better efficacy than free 8-HQN or
and DHPC respectively), the greatest pH-differential of drug release was AmB [68]. Additionally, phytochemicals (i.e., alkaloids, flavonoids,
obtained with serum at an ambient temperature of pH range 6–7.4 [53]. terpenoids, and sterols) based nanomedicines with improved PK/PD
Here, the thiolactone form of PHC was found to be more stable at acidic profiles are also being developed to prepare cost-effective drug alter­
pH. For an NP to be effective in leishmaniasis it is very important to natives with fewer side effects [66]. All these results suggest nano-based
protect it from degradation by the acidic environment and release its delivery is more effective than standard drug delivery even for natural
payload to the target site to exert potential antiparasitic activities. This compounds or newly developed antileishmanial agents. This is possible
takes into account factors like charge, hydrophobicity, nature of func­ because the solubility and bioavailability of all these compounds are
tionalized ligands affecting cellular uptake, distribution, interaction generally increased by nanoscale delivery system. A schematic presen­
with immune cells, plasma proteins, and clearance from the body [54]. tation of different NPs that is used for leishmaniasis delivery is shown in
Adsorption of plasma proteins on hydrophobic surfaces is higher [55] Fig. 2.
which in turn has an impact on the uptake, distribution, and clearance.
Organic NPs get degraded in a highly acidic lysosomal environment 4.2. Liposomes
whereas GNPs have been found to shift the pH towards the alkaline side
and reduces the lysosomal activity [56]. This indicates NP-based de­ The liposome encapsulates part of the aqueous medium with
livery can make the drug more pH tolerant. A chitosan and chondroitin dispersed hydrophilic substances whereas it accommodates hydropho­
sulfate-based NP delivery system with AmB at pH 7.4 shows its effec­ bic molecules in the bilayer and behaves as a semipermeable membrane,
tiveness in treating tegumentary leishmaniasis than standard AmB due relative to the material encapsulated in the aqueous core of the vesicles.
to improved stability and less toxicity at that pH [57]. Biopolymer of Liposomal delivery promotes superior tissue absorption, favoring drug
polycaprolactone loaded with AmB has shown better efficacy than penetration into the macrophages and retarding its clearance from the
AmBisome for cutaneous leishmaniasis (CL) at a physiological pH of 7.4 site of action with increased bioavailability. The liposome can carry both
as well as a skin-relevant pH of 5.5. It is to be noted that pH dependent water-soluble compounds and lipids and, therefore, considered drug-
delivery is directly correlated with surface charge of NPs since the delivery systems of wide application [69]. Liposomes can deliver
change in pH micro-environment affects ionization of coated functional drugs, proteins, enzymes, and genetic materials to living cells [70,71].
groups attached with NPs. Till date; no exhaustive study was performed Liposomes are more often studied for the treatment of leishmaniasis
to check the efficacy of NP-based delivery on leishmania model of compared to any other parasitic disease, mainly because Leishmania
macrophage infection at different pH. However, it is obvious that the colonizes spleen and liver macrophages, which are also responsible for
microenvironment of the phagocytes, including extreme pH, enzymatic liposome clearance in vivo. Consequently, liposome-based delivery has
or ROS-mediated degradation, and permeability of the cell membrane is minimal toxicity compared to other nano-based delivery systems. In
critical in determining the fate of internalized NPs. fact, in its early applications, liposome and its physicochemical prop­
erties were studied for macrophages, in the treatment of leishmania
4. Different NP-based drug delivery approaches in leishmaniasis infection [72]. The efficacy of various antimonial drugs such as
meglumine antimoniate (AME) or sodium stibogluconate (SSG) when
4.1. Natural and synthetic compounds/derivatives with nanoformulation encapsulated in liposomes has always shown better efficacy than the free
drug [73]. Due to drug resistance associated with free antimonials,
Through advancement in high-throughput screening of compounds, miltefosine [74] and AmB, delivery systems with liposomal form were
it is now possible to screen new compounds against the sensitive and preferred for the treatment of various types of leishmaniasis [75,76].
resistant strains of parasite compared to known antileishmanial agents. Papagiannaros and coworkers (2005) demonstrated the advantages of
The drug resistance in leishmania strains is a result of increased mem­ using liposomal miltefosine over free miltefosine with improved efficacy
brane fluidity, decreased intracellular drug accumulation, and high against miltefosine-resistant Leishmania promastigotes. Liposomal
expression of ATP-binding cassette (ABC) transporters (MDR1 and amphotericin B (L-AmB) was > 350-fold more active than AME, and
MRPA) [58]. In search of new antileishmanial agents quinoline, indolyl 2–5 times more active than non-liposomal AmB, when tested against
quinoline analogs, naphthoquinones, artemisinin, coumarins, synthetic experimental VL [77]. L-AmB eliminated 99% of the amastigotes in the
derivatives of AmB, Miltefosine, anti-cancer drug doxorubicin, etc have liver with a concomitant improvement in the AmB therapeutic index
been reported with in vitro antileishmanial activity [59–61]. Natural when tested in L. donovani-infected squirrel monkeys. Other lipid-based
and plant-derived compounds, namely andrographolide (AG), curcu­ AmB products such as AmB colloidal dispersion (Amphocil®) and the
min, bisnaphthalimidopropyl (BNIP), Oleanolic acid (OA), etc. have AmB lipid complex Amphotec® have shown less efficacy against VL than
shown strong antileishmanial activity [62,63]. The majority of these the L-AmB formulation [78] although they are less toxic and more
compounds show enhanced efficacy in nanoformulations than their efficacious than fungizone, an AmB-deoxycholate (D-AmB) formulation.
native counterpart. The chemotherapeutic applications of AG, a diter­ Ergosterol-rich liposomal AmB formulation (Kalsome™10) tested in
penoid lactone, are seriously hampered because of poor bioavailability, murine models was found to induce no hepato- or nephrotoxicity,
short plasma half-life, and unwanted tissue localization. AG nano­ leading to a significant reduction in parasite burden from liver and
particles loaded in 50:50 PLGA reduced the dose by almost 1/4th spleen [79]. Liposomes coated with sugars like galactose, mannose, and
compared to standard AG in vitro against the parasite [62]. Similarly, fucose were designed to target more specifically macrophages as these
OA which has low aqueous solubility and low permeability, when cells have receptors to recognize those glycoside residues [80,81].
administered with polymeric NPs reduced ~ 98% amastigote burden in Mannose-coated liposomal systems were more effective in vivo [82] and
the spleen of BALB/c mice, significantly higher than OA alone [63]. also the overall toxicity was reduced compared to conventional lipo­
Encapsulation of BNIP diaaminooctane (BNIPDaoct) loaded PLGA NPs somes and the free drugs [26,83].
allow the drug to be delivered orally and also reduced the parasitic Negatively charged lipids as liposomal form were found to favor the

4
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Fig. 2. A representative figure of different NPs (liposomal, niosomal, SLN, Polymeric, metallic etc.) that is used for leishmaniasis delivery.

drug-encapsulation efficiency [84], whereas cationic lipid-containing than D-AmB formulation as measured by hemolysis assay. Toxicity
positively charged liposomes had shown increased drug uptake in measured by serum creatinine and urea level was also significantly
macrophages [85]. Using PS-loaded L-AME, a high in vivo efficacy with lower for both tufstin-bearing and fufstin-free L-AmB than D-AmB.
> 130 times reduction in the dose of total antimony was observed in These studies indicate that although liposomal formulation as are fairly
parasite-infected hamsters [86]. The association of SAG with phospha­ stable and non-toxic there is a possibility of improving their delivery by
tidylcholine- stearylamine (PC-SA) liposomes had shown a significant other additional carriers. The details of liposome-based delivery of
reduction in antileishmanial activity in vivo [87]. Similarly, cationic nanoformulation against leishmaniasis are listed in Table 1.
liposomal formulation of SSG was found to be very effective for the
treatment of infection caused by SSG-sensitive and -resistant L. donovani 4.3. Niosomes
[88]. Due to preferential binding of peptides on macrophage receptors,
different peptide-modified liposomes have also shown improved dug The niosomes are lipid vesicles that contain a significant fraction of
delivery for SAG [89] and primaquine, a known antimalarial drug under non-ionic surfactants in their composition [98]. Niosomes were found to
the 8-aminoquinoline group, against leishmania parasites [90]. Due to have a higher retention capability than liposomes in vivo when the lipid
the higher success of liposomes, immunoliposomes containing lipo­ moiety was absent and the size is smaller. They are structurally similar
somes coated or bound with antibodies were also tested since macro­ to liposomes in their bilayer; however, the prepared niosomes are more
phages have receptors to bind to the Fc portion of antibodies. stable than liposomes. It can entrap both hydrophilic and lipophilic
Immunoglobulin G (IgG)-coupled liposomes [91] and liposome-bound drugs, either in an aqueous layer or in a vesicular membrane made of
antibody formulation containing doxorubicin [92] are more effective lipid material. As a result, the therapeutic effectiveness of such anti­
in killing leishmania parasites than their liposome-free counterpart. In leishmanial compounds was found to be superior to that of liposomal
short, the liposomal delivery system for leishmaniasis has huge potential antileishmanial compounds. For a long time, niosomes were considered
considering the early success of AmBisome and its extensive use to date. a viable alternative to liposomes as drug vehicles because they were less
Liposomal formulation of drugs like SAG/SSG, which are stopped due to expensive, least toxic, biodegradable, and non-immunogenic [98].
high resistance cases, may provide a new opportunity for future appli­ Itraconazole niosomes were effective against L. tropica with an IC50
cations. However, the liposomal formulations are generally of short value of 0.24 µg/ml than 0.43 µg/ml for itraconazole alone [99]. Nio­
half-life, high-cost [93] and there is a chance of leakage or fusion of somal SSG was found to be better than liposomal vesicular formulations
encapsulated drugs leading to decreased stability and toxicity in the or free drugs, against experimental murine VL by increasing drug levels
micro-environment [94,95]. Cationic liposomes have been so far known in the infected reticuloendothelial system [100]. Their data showed that
to be highly stable which if loaded with antigens acts as potent adjuvants antimony level in the liposomal form is removed much faster than nio­
to induce long-term protection against L. donovani and represents an somal form in the circulation as tested in L. donovani–infected BALB/c
alternative to DNA vaccination [96]. Stabilty of L-AmB can be increased mice models. The non-ionic surfactant vesicles with AmB have shown
by loading with tufstin, an immunomodulator of tetrapeptide sequence higher drug uptake in the lungs and skin than AmB alone thus signifi­
originated from IgG antibody. In vitro stability studies revealed that cantly reducing parasite burdens in the liver [101]. These niosomes
tuftsin-bearing AmB liposomes released ~2 fold less AmB in the serum were also used as a nebulizer indicating a different way of delivery.
of BALB/c mice, compared to tuftsin-free AmB liposomes [97]. Both Niosomes composed of either AmB, glucantine or both were prepared as
tufstin-bearing and fufstin-free L-AmB shows ~ 5 fold reduced toxicity large spherical multi-layer vesicles having a size range of 10–18 µM

5
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Table 1
Liposome-based NP delivery against leishmaniasis.
Chemotherapeutic agents/ Model of Study Mechanism of action and Advantage in using the nano-formulation Refs.
Drugs

L. donovani
Meglumine antimoniate Mice Greater drug penetration through skin [73]
Miltefosine In vivo Enhanced hepatic accumulation [74]
AmB Human Improved pharmacokinetic profile of AmB; Improved patient tolerance and reduced systemictoxicity [75,
76]
AmB PEM; Mice Improved activity due to enhanced drug uptake [78]
Ergosterol Mice No hepatotoxicity, Significant reduction in parasite burden from liver and spleen [79]
Sugars Ex vivo Macrophage specific targeting [80,
81]
Antimony Hamsters Enhanced efficacy in comparison to antimony alone [86]
SAG Ex vivo Improved drug delivery to macrophages [89]
Primaquine In vitro and in vivo Macrophage specific targeting [90]
Doxorubicin Human Enhanced killing efficacy due to targeted delivery to macrophage [91,
92]
SSG BALB/c mice Complete suppression of disease-promoting IL-10 and TGF-β, upregulation of Th1 cytokines. Increased [179]
retention of SSG within parasites.
Amphotericin-B Human Improved pharmacokinetic profile of AmB. Improved patient tolerance and reduced systemictoxicity [180]
Tuftsin decorated Hamster Increased uptake; enhanced drug tolerance [181]
IFN-g and doxorubicin- BALB/c mice Reduced levels of IL-4 but increased levels of IL-12 and iNOS.Synergistic effect of drug and liposomes. [182]
mannosylated
Artemisinin BALB/c Th1-biased immune response. Improved efficacyand no toxicity [183]
AmB Swiss mice Hamsters Up-regulation of Th-1 cytokines, iNOS, down regulation of Th-2 cytokines.Drug is morestable and less [184]
toxic
Andrographolide Hamster High macrophage delivery, effective against drug-resistant strains.Reduced parasitic burden, hepatic and [185]
renal toxicity
Cholesterol BALB/c mice Down regulation of virulence factor and Macrophage activation. Increased drug uptake [186]
L. infantum
Meglumine Antimonate Mongrel dogs and BALB/ Enhanced circulationtime. Reduction in systemic toxicity. [25]
cmice

Abbreviations: SSG, Sodium Stibogluconate; iNOS, Inducible Nitric oxide synthase; TGFβ, Transforming growth factor beta.

[102]. These glucantine and gulacntine-AmB niosomes reduce the conventional AmB without showing any toxic effects on macrophage
splenic parasite burden in L. major-infected BALB/c mice ~84% when cells [106]. To prepare an oral formulation for VL, AmB and paromo­
applied topically twice daily with 12 h intervals for 30 days. Niosomes mycin (PM) were together loaded in 2-hydroxypropyl-β-cyclodextrin
have been also evaluated as vaccine carriers for CL using purified gp63 (HPCD) modified SLNs [107]. The dual drug-loaded SLNs (DDSLNs) had
glycoprotein as antigens [103]. These gp63 proteins were encapsulated shown complete cellular internalization of SLNs within 24 h of incuba­
into niosomes and used to vaccinate C57BL/10 mice which were inoc­ tion, low cytotoxic effect without any hepatic/renal toxicity in Swiss
ulated subcutaneously twice at two weeks intervals (4 mg of pro­ albino mice. Further, the DDSLNs had shown better in vivo efficacy than
tein/mouse). When vaccinated mice were exposed to a homologous miltefosine-treated control. PM-loaded in SLNs was studied against
challenge (i.e. mice vaccinated with L. mexicana gp63 and infected with L. major-infected BALB/c mice where it showed improved effectiveness
the same parasite), significant disease resistance with ulcerated lesions than PM alone in killing the parasite by switching towards Th1 immune
was observed that began to heal. Niosome formulated with inactivated response [108]. The SLNs coated with macrophage specific ligand
whole L. major parasites were evaluated as an encapsulation model for O-palmitoyl mannan (OPM) and entrapped with AmB, showed better
vaccine candidates[98]. Although niosome-based delivery has shown parasite killing than D-AmB formulation with equivalent dose [109].
some success against leishmaniasis in animal models, till date no nio­ The organ distribution of the AmB bearing SLNs (both AmB-SLN and
somal formulation was evaluated as a replacement of L-AmB formula­ OPM-AmB-SLN) was compared with the D-AmB in albino rats. Here
tion. But, in general, the preparation of niosomes are much cheaper than OPM-coated SLNs were found to accumulate more AmB in the liver,
liposomes since niosomes use surfactants like Tween 60, Tween 80 etc spleen, and lungs, the organs which are rich in macrophages. SLNs can
which are less costly than bilayered phospholipids used in liposomes improve the drug delivery by improving the solubility of poorly
[104]. These surfactants are physically and chemically more stable than water-soluble molecules like 17-N-allylamino-17-demethoxygeldana­
phospholipids making the niosomes more stable and non-toxic than li­ mycin (17-AAG, tanespimycin) which is a known inhibitor of heat
posomes. Therefore, niosome-based delivery system may provide a shock protein 90 (Hsp90). The entrapment of 17-AAG by ~80% in SLN
suitable replacement for liposomal delivery system in the future. along with high macrophage uptake within 2 h indicated the advantage
of drug delivery with SLNs. The biggest advantage of SLN-based delivery
4.4. Solid lipid nanoparticles is its biocompatibility since the lipids used in formulation are 100%
biodegradable [110]. SLNS can be synthesized without organic solvents
Solid lipid Nanoparticles (SLNs) are nanospheres that consist of solid and they can be sterilized if required. These provide more stability and
lipids of ~ 50–1000 nm in size with a solid lipid matrix stabilized by longer storage capacity for the SLNs. The major disadvantages of SLNs
physiologically compatible emulsifiers [105]. Although the size of SLNs are the initial burst release and low drug loading capacity or expulsion
is bigger in comparison to the conventional NPs, they have been proved during the crystallization process.
to be a very efficient drug-delivery system.Chitosan-coated SLNs loaded
with AmB had shown better internalization in J774.1 cells and less 4.5. Polymer-based nanoparticles
cytotoxic effect than marketed formulations [27]. To improve AmB
bioavailability and prepare a formulation for oral delivery of AmB, a The polymeric systems are popular because they are more biocom­
vitamin B12-stearic acid (VBS) conjugate coated and SLNs encapsulated patible and biodegradable [111]. Several polymers such as poly-lactic
with AmB were fabricated. It showed an enhanced efficacy over acid (PLA), poly-glycolic acid (PGA), poly-lactide-co-glycolide (PLGA),

6
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

poly-caprolactone (PCL), and poly-cyanoacrylate (PCA) and natural molecules with improved bioavailability and reduced toxicity due to
protein polymers, such as albumin and gelatin, and polysaccharides are the ability of polymers to cross the mucosal barrier with specialized
used for the preparation of polymeric drug delivery systems [112]. The uptake mechanisms [120,121]. AmB and PLGA polymeric NPs, which
PLA, PGA, PLGA, PCL, and PCA polymers are approved by FDA for are made using E-TPGS (D-α-Tocopherol polyethylene glycol 1000 suc­
human applications due to their high biocompatibility. AmB entrapped cinate) as a stabilizer, have improved the drug’s oral bioavailability and
in PLGA NPs and stabilized by TPGS (a known p-GP efflux inhibitor) had minimize its side effects[113]. Moreover, PLGA NPs and
shown higher oral bioavailability and reduced nephrotoxicity [113,114] nano-suspensions containing AmB were also coined as an alternative of
in rats than standard fungizone. Miltefosine encapsulated PLGA–PEG cost-effective delivery compared to Fungizone and AmBisome [122]. For
NPs of a size range of 10–15 nm are more effective antileishmanial application against CL, AmB nanoencapsulation in PLGA/dimercapto­
agents[115] in the hamster model compared to standard miltefosine and succinic acid (DMSA) NPs (Nano-DMS-AmB) was developed and tested
AmB. AmB encapsulated cationic stearylamine lipid-polymer hybrid NPs against C57BL/6 mice infected with L. amazonensis. The nano-
(LPNPs) of ~ 200 nm, which had characteristics of both polymeric NPs DMS-AmB showed a greater reduction in the number of parasites than
and liposomes [116,117], had shown increased efficacy with higher standard D-AmB thereby reducing the effective dose of the nano­
macrophage uptake and rapid plasma clearance. AmB encapsulated formulation[123]. The toxicity of AmB-loaded PLGA-polyethylene gly­
sodium alginate-glycol chitosan stearate NPs (AmB-SA-GCS-NP). which col (PEG) NPs was lower compared to that of the free drug when tested
has been prepared by strong electrostatic interaction between oppositely in the macrophage model [124]. Mannose-anchored and
charged polymer and copolymer, had shown higher macrophagic uptake AmB-encapsulated PLGA NPs had shown specific targeting on macro­
in J774.1 macrophages and faster in vivo localization in tissues of liver, phage receptors thereby improving the efficacy of the drug [125].
spleen, lung, and kidney than standard AmB [118]. PS–coated gelatin Mannosylation of thiolated chitosan (TC) NPs (MTC) was found to be a
NPs with encapsulated AmB had shown higher in vivo organ distribution useful approach for improving AmB-intramacrophage localization as
in Wistar rats and also causing a more reduced parasite burden (85% vs. sustained drug release for 10 days was obtained for mannosylated NPs
50%) compared to standard AmB [119]. Polymeric NPs have also [126]. Therefore, conventional antileishmanial agents like AmB and
become useful devices for the oral administration of water-insoluble miltefosine can be delivered very efficiently using polymeric NPs both in

Table 2
Polymeric NPs-based delivery against leishmaniasis.
Chemotherapeutic agents/ drugs Model of study Mechanism of action and Advantage in using the nano-formulation Refs.

L. amazonensis
Pentamidine In vitro Controlled release; Reduced nephrotoxicity [187]
AmB In vitro Reduced IC50 against L. amazonensis; Highmacrophage uptake [188]
L. infantum
AmB (BMM-ϕ) Balb/c mice Rapid phagocytosis by macrophages and dendritic cells; Increased cytotoxic T cell [189]
response
AmB BALB/c mice Preferential accumulation in the visceral organs and increased CD-8T cell response [190]
AmB Hamsters Increased accumulation of AmB in the spleen and liver, reduced dose [191]
L. donovani
Doxorubicin Hamsters Cell cycle arrest at G1 and S phase inducing apoptosis; Increased Th1 and decreased Th2 [192]
response
Wistar rats Controlled delivery; Low cost [193]
AmB Swiss albino mice Selective macrophage targeting [125]
Rifampicin Albino rat; macrophages Greater macrophage uptake using mannose receptors [194]
AmB J774A.1; BALB/c mice Enhanced drug uptake and effective against drug-resistant strains. [126]
AmB J774A.1 Swiss albino mice Enhanced antileishmanial activity; Non- toxic [195]
AmB BMDM Equally effective but oral delivery instead of intravenous [196]
AmB Hamsters BALB/c mice Non-invasive nebulisation method; Both pulmonary and hepatic delivery [101]
Sprague–Dawley rats
Miltefosine Hamster 50% decreased dose and CD14 mediated Selective targeting [115]
Betulin J774A.1 11-fold decrease in IC50 Controlled release [197]
Curcumin and Miltefosine Hamster Synergistic effect of drugs. increased lymphocyte proliferation. [66]
Oleanolic acid BALB/c Increased solubility and bio-availability of drug; Effective against drug-resistant strains. [198]
Andrographolide – 4-fold decrease in IC50 and faster macrophage uptake [199]
Polymethyl methacrylate of Human RBC/THP-1 cells 3-fold decreased IC50 with no toxicity on human RBCs [200]
octylgallate
Polyalkylcyanoacrylate BALB/c 2-fold decreased ED50 with reduced cytotoxicity [24]
Primaquine-loaded nanoparticles J774A1 Synergistic effect of NP and primaquinone, 21-fold increase in efficacy [201]
PEG-PLA - bisnaphthalimidopropyl THP-1, J774 cells Reduced toxicity and increased liver uptake [64]
derivatives
Miltefosine In vitro and In vivo No hemolytic and cytotoxic properties [202]
Andrographolide BALB/c Sustained drug release, decrease in dosage and effective against resistant strains [203]
Curcumin J774.1 Increased solubility of curcumin and better macrophage uptake [37]
AmB BALB/c mice Rapid clearance from plasma and low distribution in kidneys with reduced toxicity [204]
Resiquimod RAW 264.7BALB/c mice 100% drug release; Increased immune response in macrophages [205]
AmB Wistar Rats, Hamster High AmB accumulation in liver and spleen with enhanced anti-leishmanial activity [119]
Artemisinin BALB/c Escalated IgG2a levels, enhanced pro-inflammatory cytokines (IFN-γ and IL-2) and [173]
suppression of Th2 cytokines (IL-10 and IL-4)
AmB Human 100-fold increased efficacy of AmB against AmB-resistant strain with poloxamer 188 [75,
206]
L. chagasi
Pentavalent antimony BALB/c mice, Hamsters Selective and improved uptake by macrophage scavenger receptors; Reduced toxicity [207]
N-(2-hydroxypropyl) THP-1 cells/ (PEMs)/ (BMMs) 19-fold increase in efficacy in vitro. [208,
methacrylamide– AmB. 209]

Abbreviations: AmB, Amphotericin B; PEM, Peritoneal macrophages; BMM, Bone marrow derived macrophages.

7
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

vitro and in vivo. The main advantages of the polymeric NPs are their agents to prepare AmB where AmB was successfully adsorbed on the
easier & faster preparation method and high drug loading with mini­ surface of biogenic AgNPs. These NPs show antileishmanial activity with
mum leakage due to the stability of polymeric NPs. More information on > 96% percent killing under visible light, indicating a better window of
Polymeric NP-based drug delivery was summarized in Table 2. using photodynamic therapy (PDT) for treating CL with skin lesions
[135]. Akhtar et al. showed that PEGylated silver doped zinc oxide (ZnO
NPs) were highly effective against leishmaniasis by developing reactive
4.6. Metallic nanoparticles oxygen species (ROS) at low concentrations, making them ideal for PDT
[136]. The biogenic AgNPs (35 nm) obtained from Anethum graveolens
Sizes, shape, high aspect ratio, and optical features of metal NPs are leaf extract enhances the activity of miltefosine on L. donovani pro­
different from their macro-scaled counterparts thus, making them ideal mastigotes and have increased biocompatibility than normal AgNPs on
to be used for drug-delivery and biosensor-based applications. In recent macrophages [137]. The biogenic GNPs (30 nm) obtained using May­
years, biological methods have been discovered to be the most suitable tenus royleanus have been shown to inhibit L. tropica promastigote’s
for the synthesis of metal NPs, as chemical and physical methods have growth in a time-dependent manner where parasite survival is directly
drawbacks [127]. Active chemical ingredients which are present in co-related with internalized GNP concentration [138]. Green synthe­
medicinal plants and plant extracts are very routinely used for the green sized titanium oxide (TiO2) NPs, synthesized using aqueous leaf extract
synthesis of metallic NPs. Since these plant products are already known of Euphorbia prostrate, showed comparatively weak leishmanicidal ac­
as antileishmanial agents, the synthesized NPs show high efficacy tivity after 24 h exposure when compared with AgNPs [139]. Novel
against the parasite-infected animal model studies [128–130] but with Au-Ag alloy NPs, which were synthesized by Dioscorea bulbifera tuber
significantly reduced cytotoxicity than chemically synthesized metallic extract, had shown a MIC value of 32 µg/ml against L. donovani pro­
NPs. Compared to AmB, the gold nanoparticles (GNP) conjugated AmB mastigotes along with anti-biofilm activity against bacteria indicating a
(GL-AmB) had shown 2.5- and a 5-fold reduction in IC50 against pro­ multipurpose application[140]. Iron oxide (Fe3O4) NPs of ~4 nm of size
mastigote and amastigote forms of the parasite, respectively [131]. synthesized by Rosmarinus officinalis leaf extract had shown an anti­
These GL-AmB NPs have reduced cytotoxicity on macrophages and were leishmanial effect on L. Major with an IC50 value of 350 µg/ml [141].
less hemolytic than standard AmB. Quercetin-functionalized GNPs were ZnO NPs synthesized from Verbena officinalis and Verbena tenuisecta
found to be highly effective against wild and resistant strains of plant leaf extracts had shown anti-leishmanial efficacy which is corre­
L. donovani parasites, with low toxicity against macrophages [128,132]. lated with the phenolic compounds present in those extracts [142].
In a recent study, we have shown that 7,8-dihydroxyflavone (DHF) Therefore, green synthesized metallic NPs synthesized have a significant
synthesized GNPs (DHF-GNP) are more antileishmanial in the amasti­ prospect in anti-leishmanial drug delivery. Among different metallic
gote model than the promastigote model [133]. The synthesized NPs, SiO2 and ZnO, are generally considered non-toxic since the bulk
DHF-GNPs of ~35 nm size kills the sensitive- and drug- (AmB and SAG) material inherently is less toxic [143]. However, there was always a
resistant parasites with equal efficacy. A diagram showing the synthesis concern over the toxicity related to GNPs and AgNPs. Production of
of NPs from the herbal extract and its application in macrophage de­ reactive oxygen species (ROS) leading to damage of cell membrane,
livery, uptake, and parasite killing is shown in Fig. 3. Adil et al. showed DNA, and protein oxidation was considered to be a general outcome of
silver nanoparticles (AgNPs) having enhanced antileishmanial activity toxicity associated with GNP and other metallic NPs [144]. The GNPs
against L. tropica by inhibiting proliferation and metabolic activity of which are administered in BALB/c mice at a dose of 8 mg/kg/week
promastigote by 1.5–3 fold respectively in dark conditions and caused severe sickness and death of mice in 21 days. These phenomena
2–6.5 fold under UV light irradiation [134]. Phytochemicals from the is observed only with 8–37 nm size of GNPs which are chemically
aqueous extract of Isatis tinctoria were used as reducing and capping

Fig. 3. A diagram showing the synthesis of NPs from herbal extract and its application in macrophage delivery, uptake and parasite killing.
(a) Giemsa staining and parasite killking data were reproduced from [255]. (b) TEM imags of macrophage NP uptake was reproduced from [256].

8
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Table 3
Metallic and miscellaneous NPs for drug-delivery against leishmaniasis.
Type of NP Chemotherapeutic Model of Study Tested Mechanism of action and Advantage in using the nano-formulation Reference
agents/ Drugs parasite

Carbon- AmB BALB/c mice L. donovani Greater solubility,increased stability at gastric pH andlow toxicity [210]
based NPs J774A.1
AmB J774A.1 No hepatic/renal toxicity of NP [211]
Hamster
Doxorubicin Hamsters High drug accumulation in parasite [212]
AmB Hamster Oral intake possible, Increase in CC50 [213]
AmB J774A.1, Wistar Increased protective proinflammatory mediators’ expression and spleen [214]
rats uptake.
Doxorubicin J774.2 Stability at lysosomal pH, high cellular uptake [215]
hamsters
Protein NPs AmB BALB/c mice L. amazonensis Reduced lesion size at infected footpad, AmB related toxicity reduced as [216]
measured by histopathological studies
Meglumine Antimonate In vitro L. major Highest activity was observed with glutaraldehyde (10 µl/ ml) [217]
Metal NPs AmB J774A.1,Wistar L. donovani Superior efficacy, desired stability and reliable safety of cost-effective [214]
rats
AmB J774A.1, Reduced cytotoxicity Increased uptake and activity [218]
Hamster
Natural compounds In vitro, in vivo Increased efficacy [127]
AgNPs In vitro Amplified anti-leishmanial effect [137]
TiO2 NPs In vitro Significant increase in G0/G1 phase of the cell cycle with a subsequent [139]
decrease in S and G2/M phases leading to growth-inhibition
AmpB In vitro, ex vivo Low cytotoxicity to macrophage cells and reduced AmpB cytotoxicity [131]
Quercetin Ex vivo Equally effective in drug-resistant strains [128]
Rhazya stricta decne-AuNPs In vitro L. tropica Effective delivery with low cytotoxicity to macrophage cells [130]

AgNPs Ex vivo Greater antimicrobial efficacy [134]


AmpB In vitro Enhanced antimicrobial activity [135]
ZnONPs In vitro Photodynamic therapy against leishmaniasis [136]
AgNPs In vivo L. major Enhanced drug-delivery [129]
Fe3O4 NPs In vitro Low toxicity [141]

synthesized (citrate or sodium borohydride) but not with 3–5 nm or known as third-generation vaccines, are of particular interest because
> 50 nm sized GNPs [145]. However, GNPs synthesized using plant they can effectively induce both CD8 + and CD4 + T cells, produce
extracts/natural products does not cause toxicity and mostly well long-lived antigens and properly folded polypeptides, etc.[149]. How­
tolerated in vitro and in vivo irrespective of the size of GNPs. This clearly ever, DNA vaccines are still in earlier phases of clinical trials.
indicates that various methods of surface modification of GNPs is related First-generation vaccine, leishmanization against CL suffers standardi­
to there toxicity and not the bulk component of the synthesized GNP. zation and safety setbacks whereas second-generation vaccines e.g. re­
Further, recent studies have shown that GNPs are gradually degraded by combinant proteins or DNA molecules (leish-111f) suffer from a lack of
NADPH oxidase in human fibroblast cells for 2–6 months [146]. This an appropriate adjuvant [150]. The second generation of vaccines
study supports the possible safety of GNP-based delivery since degraded against Leishmaniain clinical trials is of relatively low efficacy due to
particles of size < 3 nm can be easily cleared by circulation without limited CD8 + T-cell response emerging from cross-presentation [151].
causing any long-term deposition in different tissues and cells like Three types of leishmanial vaccines, Leish-F1, F2, and F3, designed at
macrophages. Table 3 summarizes metallic and other NP-based delivery the Infectious Disease Research Institute (IDRI), USA were in clinical
methods against leishmaniasis. trials [152]. LEISH-F1 which is a fusion protein made up of three
polypeptides in tandem and formulated with monophosphoryl lipid
5. Nano-based vaccine delivery for leishmaniasis A-stable emulsion (MPL-SE), showed encouraging results in phase 1 with
a shorter time to cure. The clinical trial was organized in the USA,
The dream of an effective human vaccine against leishmaniasis could Colombia, Brazil, Peru, and India. The observed safety and immunoge­
not be realized due to challenges such as failure in conferring the im­ nicity in endemic and non-endemic populations have inspired a phase II
munity, lack of suitable adjuvants, differential virulence dynamics of clinical trial with LEISH-F2 where the histidine tag of recombinant
Leishmania species, and lack of experimental human models. Addition­ protein was removed. IDRI is going for phase I trial of LEISH-F3 where
ally, the majority of the population suffering from the disease belongs to the fused peptide is composed of nucleoside hydrolase from L. donovani
poor sections of the society; therefore, low profitable recovery from the and sterol 24-c-methyltransferase from L. infantum. SLNs can serve as an
market discourages robust research initiatives to invest in vaccine-based efficient tool to synthesize and carry a leishmanial vaccine [105]. In
research for leishmaniasis. Several NP-based delivery as prophylactic 2005, a group of researchers prepared a nanovaccine delivery method in
nanovaccines for the treatment of leishmaniasis was studied [147]. A mice by loading recombinant Leishmania superoxide dismutase
scheme showing development of different nano-vaccine and its delivery (SODB1) in a chitosan NPusing the ionotropic gelation method. NPs
against animal and human models in Fig. 4. Traditional vaccines against loaded with SODB1 showed a higher cell-mediated immune response
leishmaniasis were based on live or attenuated parasites or their sub­ and higher IgG2a levels. This showed the potential of nanovaccine for
units. However, to date, only a few vaccines are under different phases the treatment of leishmaniasis. Nanoliposomes used as the nanocarriers
of the clinical trial. A summary of vaccines available for leishmania to for soluble Leishmania antigens (SLA) showed parasite clearance in the
date has been presented in Table 4. All the vaccine candidates for footpad and spleen of a mouse model injected with this formulation
leishmaniasis are categorized into three types: (i) Live leishmania; (ii) [153]. Liposomal formulations of ChimeraT (a combination of 3 leish­
Killed leishmania or parasite fractions with or without adjuvants (first manial proteins) when delivered subcutenously protected mice against
generation) and (iii) recognized leishmania molecules including re­ L. infantum infection, by reducing the parasite load in spleen, liver, bone
combinant proteins or DNA (second generation) [148]. DNA vaccines, marrow, and lymph node. The cure was associated with a stimulated

9
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Fig. 4. Development of nano-vaccine and its delivery in vivo against animal and human models.

effective adjuvant is one of the reasons for the limited efficacy. There­
Table 4
fore, a delivery system carrying both antigen and immune-stimulatory
List of vaccines developed for the treatment of leishmaniasis.
adjuvants to the antigen-presenting cells (APCs) is expected to play a
Generation Vaccine Antigen Adjuvants Refs. very significant role in enhancing efficacies. A list of nano-vaccine de­
I Leishvaccine IFLA/BR/1967/ BCG [219] livery systems prepared as potential antileishmanial vaccines is listed in
PH8 Table 5.
ALMϼ ALM Alum + BCG [220]
Nano based delivery systems (NDS) could potentially deliver target
Autoclaved L. BCG [221]
major vaccines to the site of action within the host’s body and enhance im­
Leishmune LiESP FML [222] mune responses by facilitating its absorption and uptake of antigens by
+ saponin APCs [156], prevent degradation of antigens such as peptides, proteins,
CaniLeish LiESP – [223] or oligonucleotides [157], promoting the controlled release of antigen
Attenuated Attenuated L. [224]
[158] and modulate the type of immune responses induced by the an­

vaccine donovani
Attenuated L. gentamycin [225] tigens [159]. NDS may improve the solubility of hydrophobic com­
mexicana and L. pounds in an aqueous medium to render them suitable for parenteral
major administration. Delivering antigens with the help of NPs to APCs pre­
II LEISH‑F1 LmSTI1 +LeIF MPL‑SE [](150,
sents an attractive way of activating the host immune system [148]. It
LEISH‑F2 – MPL‑SE 152)
LEISH‑F3 – GLA‑SE [152], could be used in delivering antigens and adjuvants either to enhance
[226] their uptake by APCs, [160] or generate Th1 type immune response
Leish‑Tec A2 Saponin [227] [161]. The use of nano-based vaccines could offer a stronger immuno­
SMTγ + NHμ SMT+NH GLA‑SE [228] logical response due to the delivery to the same APC [162]. Studies
KMP-11 CpG-ODN [229]
suggest that polymeric NPs when used as adjuvants could develop more
pSP IL-12 [230]
Leishmanolysin GP63 BCG, (MPL- [231] potent immunogenicity against Leishmania attributed to its physico­
TDM) chemical properties.
III ChAd63–KH ChAd63 – [232] In the case of mucosal immunization, NDS may help in the protection
Abbreviations: ALM, Autoclaved-killed L. major; SMT, sterol 24-c-methyltran­ of antigen against the gastrointestinal environment (acid and proteolytic
ferase; MPL-SE, monophosphoryl lipid A in structure stimulating Toll-like re­ enzymes), enhancement of antigen translocation to the mucosa-
ceptor; GLA, glucopyranosyl lipid A. associated lymphoid tissue, and ease of delivery via the oral or intra­
nasal route [163]. NDS-mediated delivery of antigens ensures the de­
Th1-type response in vivo by producing higher levels of IFN-γ, IL-12, and livery of both antigen and adjuvants into the same APCs which
GM-CSF cytokines [154,155]. Fig. 5. efficiently enhances activation of APCs. Additionally, encapsulated
Some encouraging efforts of developing first-generation (killed par­ formulations of immunostimulatory adjuvant into the delivery system
asites) vaccines have failed to provide convincing results in phase III restrict the systemic circulation of the adjuvant thereby preventing
trials due to standardization and safety issues whereas others are still in adverse effects [164].
earlier phases. In this regard, researchers admit that the lack of an A cationic solid–lipid nanoparticle (cSLN) formulation withA2 anti­
gen of L. donovani, and cysteine proteinases of type I (CPA) and II (CPB)

10
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Fig. 5. A graphical abstract showing delivery of different NPs against leishmania-infected macrophages involving different mechanisms of killing.

of L. infantum without its unusual C-terminal extension (CTE) showed an mice infected with L. panamensis [168]. The high encapsulation efficacy
efficient vaccine delivery system against VL [165]. Administration of the provides a tool for better antigenicity for vaccine delivery by this
pcDNA-A2-CPA-CPB(-CTE) delivered by either electroporation or cSLN method. Vaccination strategies involving live, killed, or attenuated
formulation protects BALB/c mice against L. infantum challenge asso­ (physically or genetically) leishmania parasites have produced limited
ciated with high levels of IFN-γ and lower levels of IL-10 production, success. The delivery of peptide-, protein-, or DNA-based vaccines
leading to aelevatedTh1 and reduced Th2 immune response. The ratio of although showed some promise in animal models but failed in human
IFN-γ: IL-10 induced upon re-stimulation was always significantly targets. For a suitable vaccine candidate, a strong host protective T
higher in vaccinated animals. Nitric oxide production, parasite burden, response is important [169]. However, L. donovani infection decreases
and histopathological analysis also showed a better cures for the expression of both MHC-I and MHC-II, which are responsible for
vaccine-treated animal. In a murine model, cationic liposomes formu­ effective T cell response, in macrophages and dendritic cells [170]. This
lated with a novel whole Leishmania lysate (WLL) from provides a possible explanation of the failure in vaccination strategy in
detergent-solubilized L. major promastigotes in a liposomal form, were leishmaniasis. In parallel, lack of a potential adjuvant was also reasoned
used as a vaccine for leishmaniasis [166]. The parasite burden, footpad as a major problem in vaccine delivery against leishmania. It is believed
swelling, and Th1/Th2 response were analyzed in the mice model where that nano-based vaccine delivery can replace, at least, the role of a
good efficacy was observed for vaccinated animals. In another study, a suitable adjuvant since it provides improved stability of antigen, a role
murine model of leishmaniasis was used to evaluate the role of reminiscent of most adjuvants.
liposome-polycation-DNA (LPD)-NPs containing recombinant major
surface glycoprotein of Leishmania (rgp63) in defense against leish­ 6. Possible mechanism of action of Nano-based delivery in
maniasis [167]. Reduced parasites in the spleen, the increased levels of leishmaniasis
IgG2a, minimal footpad swelling and 5-fold increased IFN-γ secretion
compared to standard SLA treatment indicated that proper adjuvants Polyamine-dependent trypanothione reductase (TR) redox system
and strong antigen-delivery systems are important for vaccine delivery and ergosterol biosynthesis pathway are two unique pathways that are
against leishmaniasis. By using a single emulsion solvent evaporation not present in the mammalian system. Therefore, any drug delivery
process, two hypothetical proteins from L. panamensis were encapsu­ methods which can target these pathways may be highly selective in
lated into PLGA particles and immune response was studied in BALB/c killing the parasite without causing any defect to its human host. The

11
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

Table 5
Nano-based vaccine delivery approaches for leishmaniasis.
Type of Antigen Model Mechanism of action References
Nanoformulation as
vaccines

Liposomes Leishmania lysate Mice Low parasite burden, footpad swelling, and Th1/Th2 response [166]
rgp63 Mice Reduced parasites in the spleen, the increased levels of IgG2a, minimal [167]
footpad swelling and 5-fold increased IFN-γ secretion
Imiquimod + SLA BALB/c mice Enhance CD-4 + and CD-8 + T cells response [233]
L. donovani antigens BALB/c mice Induction of IFN-γ and IgG2a [234]
rGP63 BALB/c mice High IgG2a/IgG1 ratio and IFN-γ production and thelowest IL-4 level [235]
Soluble leishmanial antigens BALB/c mice Strong Th1 response [236]
Recombinant L. major stress- BALB/c mice Low spleen parasite burden, high IgG2a antibody; Lower IL-4 level [237]
inducible protein 1
Recombinant LiHyR protein BALB/c mice The increased concentration of IgG2aantibody wasobserved with [238]
significantparasitic burden reduction
Recombinant glycoprotein (rgp63) BALB/c mice Induce valuable immune responseevidenced by asignificant uptickin Th-1 [167]
cells mediated immuneresponse and level of IgG2a antibody
SLA BALB/c mice Inadequate protection against Leishmaniasis [239]
Recombinant Leishmania major BALB/c mice Lowering in footpad lesion thicknessamong immunized subjects [240]
class Inuclease (rLmaCIN) withsignificantly higher levels of IgG2aand INF-γ
Freeze thawed promastigotes BALB/c mice Reduction in parasitic burden due toenhanced Th-1 cells response [241]
andassociated cytokines
Recombinant cysteine protease Syrian golden Improved anti-leishmanial responsewith increased survival time [242]
mixture Hamsters andcytokines level however immuneresponse was not adequately
evaluated
Thiolated antibody (IgG-SH) + SLA BALB/c mice Improved Cytotoxic T-cells response [243]
Virosomes Qβ-virus like α-Gal trisaccharide C57BL/6 mice The successful killing of parasites anderadication of infection [244]
particles evaluatedthrough the RT-PCR technique
Polymeric NPs PLGA Hypothetical proteins BALB/c mice High encapsulation efficiency [168]
(L. panamensis)
SLA + TLR agonists L. major Results have clearly shown improvedmacrophage activation [245]
promastigotes accompaniedby the significant reduction inpro-inflammatory cytokines

SLA + Lipophosphoglycan BALB/c mice Elevated NO, INF-γ, and IL-12 levelsdisplay 80% disease remission [246]
ininfected BALB/c
Recombinant proteins BALB/c mice Immune mediated parameters are not evaluated [168]
(rLpanUA.22; rLpanUA.27)
Recombinant proteinsChimeric BALB/c mice Improved NO levels and cell-mediated immunityfollowed by [247]
peptides (CPA and CPB) immunization
CPA Transgenic mice Enhance CD-4 + and CD-8 + T cellsresponse; Increased production of IL- [248]
12
SLA with Toll like receptor BALB/c mice Pronounced activation of cellularimmunity due to enhanced Th-1cells [248]
mediated immune responseThe levels of interleukins observed tobe
minimized(IL-6 and IL-10)
Chitosan Leishmanial SLA BALB/c mice Poor immune protection was observed [249]
PMMA pcDNA3 L. major infected BALB/c mice Improve immuneprotection wasobserved [250]
clearly evaluated throughuprise in IgG2a levels
Niosomes Autoclaved L. major BALB/c mice Amplified immune response [98]
Gp63 entrapped C57BL/10 mice Stable and controlled release [103]
AmB BALB/c Decreases in lesion size and parasite burden [251]
Solid Lipid Nanoparticles Cysteine proteinase COS-7 cells Deliver immunogenic CP genes [105]
A2 antigen BALB/c Compare electroporation and cationic solid-lipid nanoparticle (cSLN) [165]
mediated delivery to administer a DNA vaccine
rCPB C57BL/c Maximize entrapment efficiency with reduction in the particle size and its [252]
distribution
Chitosan nanoparticles Superoxide dismutase BALB/c Increased antigenicity [253]
Nanoemulsion Glucopyranosyl Lipid A (LEISH- Mice Macrophage activation [226]
F3 +GLA-SE)
Escheriosomes L. donovani promastigote soluble Hamster, BALB/c Elicit strong CD8 + cytotoxic T lymphocyte (CTL) response [254]
antigens

Abbreviations: SLA, Soluble Leishmania antigen; IgG, Immunoglobulin; IL, Interleukin; CTL, Cytotoxic T lymphocytes

crystal structure of L. infantum TR in complex with NADPH and Ag(0) trypanathione reductase (TR) activity and subsequently reduced thiol
was solved at 3.3 Å resolution [171]. Ag binds with high affinity to the content was found to be decreased under similar conditions. This in­
catalytic triad composed of Cys52, Cys57, and His461 residues of dicates that conjugate GL-AmB acts on both targets involving ergosterol
L. infantum TR thereby causing parasite death. Further in vitro assay biosynthesis and the TR redox pathway. Although, GL-AmB causes
with Ag showed that Ag can inhibit TR activity in nanomolar concen­ increased ROS necrosis, instead of apoptosis, was observed as evidenced
tration and it can be more effective than antimony. Green synthesized by DNA fragmentation, FACS, and lactate dehydrogenase (LDH) leakage
AgNPs and TiO2 NPs kills the parasite by cell cycle arrest at G0/G1 phase assay. The drugs which generate ROS are preferred leishmanicidal
thereby reducing the promastigotes in the S and G2/M phases of the cell agents, as Leishmania has to survive under the high oxidative stress of
cycle [139]. Further, increased ROS production leading to increased macrophages where they primarily infect and reside [136]. Metallic NPs
apoptosis and necrosis was observed. GNP conjugated with AmB which produce ROS are always effective against many pathogenic mi­
(GL-AmB) kills the parasite by decreasing the ergosterol content ~2 crobes albeit with a problem related to cytotoxicity [172]. The NPs
times more than the same equivalent of free AmB. In parallel, entrapped with natural compounds, phytochemicals or flavonoids,

12
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

therefore, showed increased uptake and less cytotoxicity in shown by various studies.
macrophage-infected parasites although generally, they do not show
enhanced effect in vitro against promastigotes than their naked coun­ CRediT authorship contribution statement
terpart [66,68,173]. Green synthesized GNPs were found to deposit in­
side the macrophages within hours of treatment thereby conferring their PP, PK, and DM designed the concept; PP, PK, SK, VKR, VK, SRP and
effectiveness against drug-resistant strains of L. donovani [130,174,175]. DM collected information and data. PP, PK,SK,VKR, and DM written the
GNP functionalized with DHF showed that inhibition of arginase activity manuscript; PP, PK, UM, VR and DM analyzed the manuscript. All au­
of polyamine biosynthesis pathway of the parasite is the major cause of thors read and approved the final manuscript.
death [133]. DHF-GNP was found to be a more potent inhibitor of
arginase than DHF alone but DHF-GNP was more biocompatible than Funding
citrate-reduced GNPs. Interestingly, both DHF and DHF-GNP showed
inhibition of drug transporter, MDR1, thereby making it effective The funding for this research work was provided by the Ministry of
against drug-resistant strains of the parasite. This study further confirms Chemicals and Fertilizers, Government of India.
that the toxicity of metallic NPs can be reduced by molecules used in
surface functionalization. The liposome or polymeric NP-based delivery
of AmB has always reduced cytotoxicity than free AmB, thereby, Competing Interests
allowing increased dosage for application in animal models [77–79]. In
the macrophage model, NP-based delivery with reduced parasite count The authors report no conflicts of interest in this work.
always comes with a common observation which is concomitant with
increased Th1 and decreased Th2 response [27,79,88,131]. Acknowledgment
Up-regulation of proinflammatory cytokines such as IL-12 and IFN-γ
(Th1 markers) and limiting the expression of IL-10 (Th2 markers) along We acknowledge the financial assistance in research funding to Dr.
with Th1 cytokines-mediated increased expression of inducible nitric Debabrata Mandal and Ph.D.fellowship to Pragya Prasanna, Vinod
oxide synthase (iNOS) was found to be the most common mechanism for Kumar Rajana, Saurabh Kumar, Vishnu Kant and Surendra Rajit Prasad
clearing parasites in vivo in leishmaniasis [176]. In nano-based vaccine from the Ministry of Chemicals and Fertilizers, Government of India. Dr.
delivery system increased stability of antigen [158]. The higher ratio of Prakash Kumar was assisted by a Post doctoral research fellowship from
Th1/Th2 cytokine response [166].and increased IgG2a/IgG1 ratio for the Ministry of Chemicals and Fertilizers, Government of India.
polysaccharide encapsulated NPs [167] was observed for BALB/c mice
which are vaccinated. References

7. Conclusion [1] P. Mohanpuria, N. Rana, Biosynthesis of nanoparticles: technological concepts


and future applications Biosynthesis of nanoparticles: technological concepts and
future applications, J. Nanopart. Res 10 (2008) 507–517.
Macrophages (~2000 nm) composed of DNA (~2.5 nm) and pro­ [2] O.V. Salata, Applications of nanoparticles in biology and medicine,
teins (~1–20 nm) can be considered very efficient targets in leishman­ J. Nanobiotechnol. 2 (2004) 3.
iasis for NP-based drug delivery due to their different sizes in the nm [3] A.K. Saim, F.N. Kumah, M.N. Oppong, Extracellular and intracellular synthesis of
gold and silver nanoparticles by living plants: a review, Nanotechnol. Environ.
range. However, very little progress was made for efficient use of NP- Eng. 6 (2021) 1–11.
based drugs in leishmaniasis, except the liposomal formulation of [4] H.N. Abdelhamid, Self-decontaminating antimicrobial textiles, in: Antimicrob.
AmB which was FDA approved in the year 1997. The toxicity of most Text. from Nat. Resour., Elsevier, 2021, pp. 259–294.
[5] H.N. Abdelhamid, H.F. Wu, Proteomics analysis of the mode of antibacterial
chemotherapeutic drugs, drug resistance, high cost of liposomal for­ action of nanoparticles and their interactions with proteins, TrAC Trends Anal.
mulations, and consequent lack of interest of private firms due to low Chem. 65 (2015) 30–46.
benefit in treating a poorer class of people in 3rd world countries had [6] A. de Souza, D.S.S. Marins, S.L. Mathias, L.M. Monteiro, M.N. Yukuyama, C.
B. Scarim, R. Löbenberg, N.A. Bou-Chacra, Promising nanotherapy in treating
created a big hole in eradicating the disease by 2020 as proposed by
leishmaniasis, Int. J. Pharm. 547 (2018) 421–431.
WHO [8]. Post Kala-Azar dermal leishmaniasis (PKDL) which is usually [7] M. Akbari, A. Oryan, G. Hatam, Application of nanotechnology in treatment of
a sequel of VL and the emergence of HIV-VL co-infections generates a leishmaniasis: a review, Acta Trop. 172 (2017) 86–90.
[8] S. Burza, S.L. Croft, M. Boelaert, Leishmaniasis, Lancet 392 (2018) 951–970.
new sociological problem related to leishmaniasis treatment. Since
[9] M. Vannier-Santos, A. Martiny, W. Souza, Cell biology of Leishmania spp.:
1995, more than 50 nano pharmaceuticals have received FDA approval invading and evading, Curr. Pharm. Des. 8 (2005) 297–318.
and are currently available for clinical use for various diseases including [10] C.P. Thakur, G.P. Sinha, A.K. Pandey, N. Kumar, P. Kumar, S.M. Hassan,
different forms of cancer, multiple sclerosis, SCID, and even for diseases S. Narain, R.K. Roy, Do the diminishing efficacy and increasing toxicity of sodium
stibogluconate in the treatment of visceral leishmaniasis in Bihar, India, justify its
related to iron deficiency [177]. However, no progress was made so far continued use as a first line drug? An observational study of 80 cases, Ann. Trop.
for VL and CL. These nanodrugs are typically administered orally, Med. Parasitol. 92 (1998) 561–569.
intravenously, and less frequently transdermally and in different forms [11] S. Sundar, M. Rai, Laboratory diagnosis of visceral leishmaniasis, Clin. Diagn.
Lab. Immunol. 9 (2002) 951–958.
(Polymeric, liposomal, metallic, nanocrystals, etc.) very successfully. [12] S. Sundar, P.K. Sinha, M. Rai, D.K. Verma, K. Nawin, S. Alam, J. Chakravarty,
Currently, there are also many formulations being developed for other M. Vaillant, N. Verma, K. Pandey, P. Kumari, C.S. Lal, R. Arora, B. Sharma,
indications, including metabolic disorders, autoimmune conditions, S. Ellis, N. Strub-Wourgaft, M. Balasegaram, P. Olliaro, P. Das, F. Modabber,
Comparison of short-course multidrug treatment with standard therapy for
anesthesia, ophthalmic conditions, neurological and psychiatric dis­ visceral leishmaniasis in India: an open-label, non-inferiority, randomised
eases, and others [178]. It indicates the possible safety and advantage of controlled trial, Lancet 377 (2011) 477–486.
targeted drug delivery of nanoformulation which is a major problem in [13] S. Mistro, B. Gomes, L. Rosa, L. Miranda, M. Camargo, R. Badaró, Cost-
effectiveness of liposomal amphotericin B in hospitalised patients with
conventional delivery. Most of the nanodrugs approved to date have
mucocutaneous leishmaniasis, Trop. Med. Int. Heal. 22 (2017) 1569–1578.
shown reduced toxicity rather than improved efficacy compared to [14] B. Monge-Maillo, R. López-Vélez, L.D. Saravolatz, Miltefosine for visceral and
conventional formulations. The most obvious advantage of NP-based cutaneous leishmaniasis: drug characteristics and evidence-based treatment
recommendations, Clin. Infect. Dis. 60 (2015) 1398–1404.
delivery is the customization of the delivery system with different
[15] J. Alvar, I.D. Vélez, C. Bern, M. Herrero, P. Desjeux, J. Cano, J. Jannin, M. de
sizes, shapes, and cargo thereby providing targeted and sustained drug Boer, Leishmaniasis worldwide and global estimates of its incidence, PLoS One 7
release at the desired tissues and organs. Therefore, nanoscale- drug (2012) 35671.
delivery may become a suitable method for treatments of diseases like [16] P. Desjeux, Leishmaniasis: current situation and new perspectives, Comp.
Immunol. Microbiol. Infect. Dis. 27 (2004) 305–318.
leishmaniasis where the mechanism of action and efficacy in reducing [17] H.W. Murray, J.D. Berman, C.R. Davies, N.G. Saravia, Advances in leishmaniasis,
the parasite burden, in vivo, by different nano-formulations was already in: Lancet, Elsevier B.V, 2005, pp. 1561–1577.

13
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

[18] C.R. Alving, E.A. Steck, W.L. Chapman, V.B. Waits, L.D. Hendricks, G.M. Swartz, [47] T.D. Heath, N.G. Lopez, D. Papahadjopoulos, The effects of liposome size and
W.L. Hanson, Therapy of leishmaniasis: superior efficacies of liposome surface charge on liposome-mediated delivery of methotrexate-γ-aspartate to cells
encapsulated drugs, Proc. Natl. Acad. Sci. U. S. A. 75 (1978) 2959–2963. in vitro, BBA Biomembr. 820 (1985) 74–84.
[19] O.P. Singh, M.R. Gedda, S.L. Mudavath, O.N. Srivastava, S. Sundar, Envisioning [48] Y. Yao, P.E. Saw, Y. Nie, P.P. Wong, L. Jiang, X. Ye, J. Chen, T. Ding, L. Xu,
the innovations in nanomedicine to combat visceral leishmaniasis: for future H. Yao, H. Hu, X. Xu, Multifunctional sharp pH-responsive nanoparticles for
theranostic application, Nanomedicine 14 (2019) 1911–1927. targeted drug delivery and effective breast cancer therapy, J. Mater. Chem. B 7
[20] A.Z. Wilczewska, K. Niemirowicz, K.H. Markiewicz, H. Car, Nanoparticles as drug (2019) 576–585.
delivery systems, Pharmacol. Rep. 64 (2012) 1020–1037. [49] L. Palanikumar, S. Al-Hosani, M. Kalmouni, V.P. Nguyen, L. Ali, R. Pasricha, F.
[21] E. Blanco, H. Shen, M. Ferrari, Principles of nanoparticle design for overcoming N. Barrera, M. Magzoub, pH-responsive high stability polymeric nanoparticles for
biological barriers to drug delivery, Nat. Biotechnol. 33 (2015) 941–951. targeted delivery of anticancer therapeutics, Commun. Biol. 3 (2020) 95.
[22] H. Nehoff, N.N. Parayath, L. Domanovitch, S. Taurin, K. Greish, Nanomedicine for [50] Y. Zhang, J. Ye, N. Hosseini-Nassab, A. Flores, I. Kalashnikova, S.L. Paluri,
drug targeting: strategies beyond the enhanced permeability and retention effect, M. Lotfi, N.J. Leeper, B.R. Smith, Macrophage-targeted single walled carbon
Int. J. Nanomed. 9 (2014) 2539–2555. nanotubes stimulate phagocytosis via pH-dependent drug release, Nano Res. 14
[23] N. Bruni, B. Stella, L. Giraudo, C. Della Pepa, D. Gastaldi, F. Dosio, (2021) 762–769.
Nanostructured delivery systems with improved leishmanicidal activity: a critical [51] J.B. Dressman, R.R. Berardi, L.C. Dermentzoglou, T.L. Russell, S.P. Schmaltz, J.
review, Int. J. Nanomed. 12 (2017) 5289–5311. L. Barnett, K.M. Jarvenpaa, Upper Gastrointestinal (GI) pH in young, healthy men
[24] A. Nan, S.L. Croft, V. Yardley, H. Ghandehari, Targetable water-soluble polymer- and women, Pharm. Res. 7 (1990) 756–761.
drug conjugates for the treatment of visceral leishmaniasis, J. Control. Release 94 [52] T.T. Kararli, Comparison of the gastrointestinal anatomy, physiology, and
(2004) 115–127. biochemistry of humans and commonly used laboratory animals, Biopharm. Drug
[25] E.G. Azevedo, R.R. Ribeiro, S.M. Da Silva, C.S. Ferreira, L.E. De Souza, A.A. Dispos. 16 (1995) 351–380.
F. Ferreira, R.A. De Oliveira, E. Castro, C. Demicheli, S.A. Rezende, F. Frézard, [53] B. Conti, C. Bucolo, C. Giannavola, G. Puglisi, P. Giunchedi, U. Conte,
Mixed formulation of conventional and pegylated liposomes as a novel drug Biodegradable microspheres for the intravitreal administration of acyclovir: In
delivery strategy for improved treatment of visceral leishmaniasis, Expert Opin. vitro/in vivo evaluation, Eur. J. Pharm. Sci. 5 (1997) 287–293.
Drug Deliv. 11 (2014) 1551–1560. [54] P. Aggarwal, J.B. Hall, C.B. McLeland, M.A. Dobrovolskaia, S.E. McNeil,
[26] G. Banerjee, G. Nandi, S.B. Mahato, A. Pakrashi, M.K. Basu, Drug delivery system: Nanoparticle interaction with plasma proteins as it relates to particle
targeting of pentamidines to specific sites using sugar grafted liposomes, biodistribution, biocompatibility and therapeutic efficacy, Adv. Drug Deliv. Rev.
J. Antimicrob. Chemother. 38 (1) (1996) 145–150. 61 (2009) 428–437.
[27] V. Jain, A. Gupta, V.K. Pawar, S. Asthana, A.K. Jaiswal, A. Dube, M.K. Chourasia, [55] T.M. Göppert, R.H. Müller, Adsorption kinetics of plasma proteins on solid lipid
Chitosan-assisted immunotherapy for intervention of experimental leishmaniasis nanoparticles for drug targeting, Int. J. Pharm. 302 (2005) 172–186.
via amphotericin B-loaded solid lipid nanoparticles, Appl. Biochem. Biotechnol. [56] B.B. Manshian, S. Pokhrel, L. Mädler, S.J. Soenen, The impact of nanoparticle-
174 (2014) 1309–1330. driven lysosomal alkalinization on cellular functionality, J. Nanobiotechnol. 16
[28] M. Yasinzai, M. Khan, A. Nadhman, G. Shahnaz, Drug resistance in leishmaniasis: (2018) 85.
current drug-delivery systems and future perspectives, Future Med. Chem. 5 [57] T.G. Ribeiro, J.R. Franca, L.L. Fuscaldi, et al., An optimized nanoparticle delivery
(2013) 1877–1888. system based on chitosan and chondroitin sulfate molecules reduces the toxicity
[29] M.B. Yatvin, W. Kreutz, B.A. Horwitz, M. Shinitzky, pH-sensitive liposomes: of amphotericin B and is effective in treating tegumentary leishmaniasis, Int. J.
possible clinical implications, Sci. 80 (210) (1980) 1253–1255. Nanomed. 9 (2014) 5341–5353.
[30] E. Yuba, N. Tajima, Y. Yoshizaki, A. Harada, H. Hayashi, K. Kono, Dextran [58] A. Klokouzas, S. Shahi, S.B. Hladky, M.A. Barrand, H.W. Van, Veen, ABC
derivative-based pH-sensitive liposomes for cancer immunotherapy, Biomaterials transporters and drug resistance in parasitic protozoa, Int. J. Antimicrob. Agents
35 (2014) 3091–3101. 22 (2003) 301–317.
[31] S. Cerritelli, D. Velluto, J.A. Hubbell, PEG-SS-PPS: reduction-sensitive disulfide [59] T. Silva, M.A. Abengózar, M. Fernandez-Reyes, D. Andreu, K. Nazmi, J.G.
block copolymer vesicles for intracellular drug delivery, Biomacromolecules 8 M. Bolscher, M. Bastos, L. Rivas, Enhanced leishmanicidal activity of
(2007) 1966–1972. cryptopeptide chimeras from the active N1 domain of bovine lactoferrin, Amino
[32] S. George, S. Lin, Z. Ji, C.R. Thomas, et al., Surface defects on plate-shaped silver Acids 43 (2012) 2265–2277.
nanoparticles contribute to its hazard potential in a fish gill cell line and zebrafish [60] A.K. Marr, B.S. McGwire, W.R. McMaster, Modes of action of Leishmanicidal
embryos, ACS Nano 6 (2012) 3745–3759. antimicrobial peptides, Future Microbiol. 7 (2012) 1047–1059.
[33] J. Hua, M.G. Vijver, M.K. Richardson, et al., Particle-specific toxic effects of [61] A. Fortin, S. Hendrickx, V. Yardley, P. Cos, H. Jansen, L. Maes, Efficacy and
differently shaped zinc oxide nanoparticles to zebrafish embryos (Danio rerio), tolerability of oleylphosphocholine (OLPC) in a laboratory model of visceral
Environ. Toxicol. Chem. 33 (2014) 2859–2868. leishmaniasis, J. Antimicrob. Chemother. 67 (2012) 2707–2712.
[34] T. Borel, C.M. Sabliov, Nanodelivery of bioactive components for food [62] P. Roy, S. Das, T. Bera, S. Mondol, A. Mukherjee, Andrographolide nanoparticles
applications: types of delivery systems, properties, and their effect on ADME in leishmaniasis: characterization and in vitro evaluations, Int. J. Nanomed. 5
profiles and toxicity of nanoparticles, Annu. Rev. Food Sci. Technol. 5 (2014) (2010) 1113–1121.
197–213. [63] S. Ghosh, N. Kar, T. Bera, Oleanolic acid loaded poly lactic co- glycolic acid-
[35] M. Basu, S. Lala, Macrophage specific drug delivery in experimental vitamin E TPGS nanoparticles for the treatment of Leishmania donovani infected
leishmaniasis, Curr. Mol. Med. 4 (2005) 681–689. visceral leishmaniasis, Int. J. Biol. Macromol. 93 (2016) 961–970.
[36] X. Xie, J. Liao, X. Shao, Q. Li, Y. Lin, The effect of shape on cellular uptake of gold [64] S.A. Costa Lima, M. Resende, R. Silvestre, J. Tavares, A. Ouaissi, P.K.T. Lin,
nanoparticles in the forms of stars, rods, and triangles, Sci. Rep. 7 (2017) 1–9. A. Cordeiro-Da-Silva, Characterization and evaluation of BNIPDaoct-loaded PLGA
[37] K. Nambara, K. Niikura, H. Mitomo, et al., Reverse size dependences of the nanoparticles for visceral leishmaniasis: In vitro and in vivo studies,
cellular uptake of triangular and spherical gold nanoparticles, Langmuir 32 Nanomedicine 7 (2012) 1839–1849.
(2016) 12559–12567. [65] M.Y. Want, M. Islamuddin, G. Chouhan, A.K. Dasgupta, A.P. Chattopadhyay,
[38] Y. Jiang, S. Huo, T. Mizuhara, R. Das, et al., The interplay of size and surface F. Afrin, A new approach for the delivery of artemisinin: formulation,
functionality on the cellular uptake of sub-10 nm gold nanoparticles, ACS Nano 9 characterization, and ex-vivo antileishmanial studies, J. Colloid Interface Sci. 432
(2015) 9986–9993. (2014) 258–269.
[39] M.M. Arnida, A. Janát-Amsbury, C.M. Ray, H. Peterson, Ghandehari, geometry [66] B. Tiwari, R. Pahuja, P. Kumar, S.K. Rath, K.C. Gupta, N. Goyal, Nanotized
and surface characteristics of gold nanoparticles influence their biodistribution curcumin and miltefosine, a potential combination for treatment of experimental
and uptake by macrophages, Eur. J. Pharm. Biopharm. 77 (2011) 417–423. visceral leishmaniasis, Antimicrob. Agents Chemother. 61 (3) (2017), e01169.
[40] A. Gessner, A. Lieske, B.R. Paulke, R.H. Müller, Influence of surface charge [67] L. Paloque, P. Verhaeghe, M. Casanova, C. Castera-Ducros, A. Dumètre,
density on protein adsorption on polymeric nanoparticles: analysis by two- L. Mbatchi, S. Hutter, M. Kraiem-M’Rabet, M. Laget, V. Remusat, S. Rault,
dimensional electrophoresis, Eur. J. Pharm. Biopharm. 54 (2002) 165–170. P. Rathelot, N. Azas, P. Vanelle, Discovery of a new antileishmanial hit in 8-
[41] E. Fröhlich, The role of surface charge in cellular uptake and cytotoxicity of nitroquinoline series, Eur. J. Med. Chem. 54 (2012) 75–86.
medical nanoparticles, Int. J. Nanomed. 7 (2012) 5577–5591. [68] M. Costa Duarte, L.M. dos Reis, Lage et al., An effective in vitro and in vivo
[42] V.A. Fadok, A. De Cathelineau, et al., Loss of phospholipid asymmetry and surface antileishmanial activity and mechanism of action of 8-hydroxyquinoline against
exposure of phosphatidylserine is required for phagocytosis of apoptotic cells by Leishmania species causing visceral and tegumentary leishmaniasis, Vet.
macrophages and fibroblasts, J. Biol. Chem. 276 (2001) 1071–1077. Parasitol. 217 (2016) 81–88.
[43] J.M. De Freitas Balanco, M.E. Costa Moreira, et al., Apoptotic mimicry by an [69] W.E. Bawarski, E. Chidlowsky, D.J. Bharali, S.A. Mousa, Emerging
obligate intracellular parasite downregulates macrophage microbicidal activity, nanopharmaceuticals, Nanomedicine Nanotechnology, Biol. Med. 4 (2008)
Curr. Biol. 11 (2001) 1870–1873. 273–282.
[44] S. Jeon, J. Clavadetscher, D.K. Lee, S.V. Chankeshwara, M. Bradley, W.S. Cho, [70] A. Akbarzadeh, R. Rezaei-Sadabady, S. Davaran, S.W. Joo, N. Zarghami,
Surface charge-dependent cellular uptake of polystyrene nanoparticles, Y. Hanifehpour, M. Samiei, M. Kouhi, K. Nejati-Koshki, Liposome: classification,
Nanomaterials 8 (12) (2018) 1028. preparation, and applications, Nanoscale Res. Lett. 8 (2013) 102.
[45] D.E. Owens, N.A. Peppas, Opsonization, biodistribution, and pharmacokinetics of [71] C. Kelly, C. Jefferies, S.-A. Cryan, Targeted liposomal drug delivery to monocytes
polymeric nanoparticles, Int. J. Pharm. 307 (2006) 93–102. and macrophages, J. Drug Deliv. 2011 (2011) 1–11.
[46] G. Sanità, B. Carrese, A. Lamberti, Nanoparticle surface functionalization: how to [72] M. Owais, C. Gupta, Targeted drug delivery to macrophages in parasitic
improve biocompatibility and cellular internalization, Front. Mol. Biosci. 7 infections, Curr. Drug Deliv. 2 (2005) 311–318.
(2020), 587012. [73] S.A. Moosavian Kalat, A. Khamesipour, N. Bavarsad, M. Fallah,
Z. Khashyarmanesha, E. Feizi, K. Neghabi, A. Abbasi, M.R. Jaafari, Use of topical

14
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

liposomes containing meglumine antimoniate (Glucantime) for the treatment of [101] M. Alsaadi, J.L. Italia, A.B. Mullen, et al., The efficacy of aerosol treatment with
L. major lesion in BALB/c mice, Exp. Parasitol. 143 (2014) 5–10. non-ionic surfactant vesicles containing amphotericin B in rodent models of
[74] A. Papagiannaros, C. Bories, C. Demetzos, P.M. Loiseau, Antileishmanial and leishmaniasis and pulmonary aspergillosis infection, J. Control. Release 160
trypanocidal activities of new miltefosine liposomal formulations, Biomed. (2012) 685–691.
Pharmacother. 59 (2005) 545–550. [102] M. Mostafavi, I. Sharifi, S. Farajzadeh, P. Khazaeli, H. Sharifi, E. Pourseyedi,
[75] P. Minodier, K. Retornaz, A. Horelt, J.M. Garnier, Liposomal amphotericin B in S. Kakooei, M. Bamorovat, A. Keyhani, M.H. Parizi, A. Khosravi, A. Khamesipour,
the treatment of visceral leishmaniasis in immunocompetent patients, Fundam. Niosomal formulation of amphotericin B alone and in combination with
Clin. Pharmacol. 17 (2003) 183–188. glucantime: In vitro and in vivo leishmanicidal effects, Biomed. Pharmacother.
[76] P.R.L. MacHado, M.E.A. Rosa, L.H. Guimarães, F.V.O. Prates, A. Queiroz, 116 (2019), 108942.
A. Schriefer, E.M. Carvalho, Treatment of disseminated leishmaniasis with [103] C.M. Lezama-Dávila, Vaccination of C57BL/10 mice against cutaneous
liposomal amphotericin B, Clin. Infect. Dis. 61 (2015) 945–949. leishmaniasis. Use of purified gp63 encapsulated into niosomes surfactants
[77] P.V. Bodhe, R.N. Kotwani, B.G. Kirodian, A.V. Pathare, A.K. Pandey, C.P. Thakur, vesicles: a novel approach, Mem. Inst. Oswaldo Cruz. 94 (1999) 67–70.
N.A. Kshirsagar, Dose-ranging studies on liposomal amphotericin B (L-AMP-LRC- [104] M. Basu, S. Lala, Macrophage specific drug delivery in experimental
1) in the treatment of visceral leishmaniasis, Trans. R. Soc. Trop. Med. Hyg. 93 leishmaniasis, Curr. Mol. Med. 4 (6) (2005) 681–689.
(1999) 314–318. [105] D. Doroud, A.V. Vatanara, F. Zahedifard, et al., Cationic solid lipid nanoparticles
[78] V. Yardley, S.L. Croft, A comparison of the activities of three amphotericin B lipid loaded by cysteine proteinase genes as a novel anti-leishmaniasis DNA vaccine
formulations against experimental visceral and cutaneous leishmaniasis, Int. J. delivery system: characterization and in vitro evaluations, J. Pharm. Pharm. Sci.
Antimicrob. Agents 13 (2000) 243–248. 13 (2010) 320–335.
[79] M. Asad, P. Bhattacharya, A. Banerjee, N. Ali, Therapeutic and [106] A. Singh, G. Yadagiri, S. Parvez, O.P. Singh, A. Verma, S. Sundar, S.L. Mudavath,
immunomodulatory activities of short-course treatment of murine visceral Formulation, characterization and in vitro anti-leishmanial evaluation of
leishmaniasis with KALSOMETM10, a new liposomal amphotericin B, BMC Infect. amphotericin B loaded solid lipid nanoparticles coated with vitamin B12-stearic
Dis. 15 (2015) 188. acid conjugate, Mater. Sci. Eng. C 117 (2020), 111279.
[80] K. Jain, P. Kesharwani, U. Gupta, N.K. Jain, A review of glycosylated carriers for [107] S. Parvez, G. Yadagiri, M.R. Gedda, A. Singh, O.P. Singh, A. Verma, S. Sundar, S.
drug delivery, Biomaterials 33 (2012) 4166–4186. L. Mudavath, Modified solid lipid nanoparticles encapsulated with Amphotericin
[81] V. Ortega, S. Giorgio, E. de Paula, Liposomal formulations in the pharmacological B and Paromomycin: an effective oral combination against experimental murine
treatment of leishmaniasis: a review, J. Liposome Res. 27 (2017) 234–248. visceral leishmaniasis, Sci. Rep. 10 (2020) 12243.
[82] B. Raay, S. Medda, S. Mukhopadhyay, M.K. Basu, Targeting of piperine [108] M. Heidari-Kharaji, T. Taheri, D. Doroud, S. Habibzadeh, S. Rafati, Solid lipid
intercalated in mannose-coated liposomes in experimental leishmaniasis, Indian, nanoparticle loaded with paromomycin: in vivo efficacy against Leishmania
J. Biochem. Biophys. 36 (1999) 248–251. tropica infection in BALB/c mice model, Appl. Microbiol. Biotechnol. 100 (2016)
[83] P.G. Bray, M.P. Barrett, S.A. Ward, H.P. De Koning, Pentamidine uptake and 7051–7060.
resistance in pathogenic protozoa: Past, present and future, Trends Parasitol. 19 [109] S. Gupta, A. Dube, S.P. Vyas, Development and characterization of amphotericin
(2003) 232–239. B loaded solid lipid nanoparticles against experimental visceral leishmaniasis,
[84] A. Momeni, M. Rasoolian, A. Momeni, A. Navaei, S. Emami, Z. Shaker, Pharm. Nanotechnol. 1 (2013) 54–67.
M. Mohebali, A. Khoshdel, Development of liposomes loaded with anti- [110] Y. Zhai, G. Zhai, Advances in lipid-based colloid systems as drug carrier for topic
leishmanial drugs for the treatment of cutaneous leishmaniasis, J. Liposome Res. delivery, J. Control. Release 193 (2014) 90–99.
23 (2013) 134–144. [111] H.K. Makadia, S.J. Siegel, Poly Lactic-co-Glycolic Acid (PLGA) as biodegradable
[85] P. Qi, M. Cao, L. Song, C. Chen, M. Liu, N. Li, D. Wu, J. Peng, G. Hu, J. Zhao, The controlled drug delivery carrier, in: Polymers, 3, Basel, 2011, pp. 1377–1397.
biological activity of cationic liposomes in drug delivery and toxicity test in [112] Z. Liu, Y. Jiao, Y. Wang, C. Zhou, Z. Zhang, Polysaccharides-based nanoparticles
animal models, Environ. Toxicol. Pharmacol. 47 (2016) 159–164. as drug delivery systems, Adv. Drug Deliv. Rev. 60 (2008) 1650–1662.
[86] A.G. Tempone, Targeting Leishmania (L.) chagasi amastigotes through [113] J.L. Italia, M.M. Yahya, D. Singh, M.N.V. Ravi, Kumar, Biodegradable
macrophage scavenger receptors: the use of drugs entrapped in liposomes nanoparticles improve oral bioavailability of amphotericin B and show reduced
containing phosphatidylserine, J. Antimicrob. Chemother. 54 (2004) 60–68. nephrotoxicity compared to intravenous fungizone®, Pharm. Res. 26 (2009)
[87] S. Pal, R. Ravindran, N. Ali, Combination therapy using sodium antimony 1324–1331.
gluconate in stearylamine-bearing liposomes against established and chronic [114] Z. Zhang, S. Tan, S.S. Feng, Vitamin E TPGS as a molecular biomaterial for drug
Leishmania donovani infection in BALB/c mice, Antimicrob. Agents Chemother. delivery, Biomaterials 33 (2012) 4889–4906.
48 (2004) 3591–3593. [115] R. Kumar, G.C. Sahoo, K. Pandey, V.N.R. Das, R.K. Topno, M.Y. Ansari, P. Das,
[88] R. Sinha, J. Roychoudhury, P. Palit, N. Ali, Cationic liposomal sodium Development of PLGA-PEG encapsulated miltefosine based drug delivery system
stibogluconate (ssg), a potent therapeutic tool for treatment of infection by ssg- against visceral leishmaniasis, Mater. Sci. Eng. C 59 (2016) 748–753.
sensitive and resistant leishmania donovani, Antimicrob. Agents Chemother. 59 [116] P.K. Gupta, S. Asthana, A.K. Jaiswal, V. Kumar, A.K. Verma, P. Shukla,
(2015) 334–354. P. Dwivedi, A. Dube, P.R. Mishra, Exploitation of lectinized lipo-polymerosome
[89] P.Y. Guru, A.K. Agrawal, U.K. Singha, A. Singhal, C.M. Gupta, Drug targeting in encapsulated amphotericin b to target macrophages for effective chemotherapy of
Leishmania donovani infections using tuftsin-bearing liposomes as drug vehicles, visceral leishmaniasis, Bioconjug. Chem. 25 (2014) 1091–1102.
FEBS Lett. 245 (1989) 204–208. [117] S. Asthana, A.K. Jaiswal, et al., Th-1 biased immunomodulation and synergistic
[90] G. Banerjee, S. Medda, M.K. Basu, A novel peptide-grafted liposomal delivery antileishmanial activity of stable cationic lipid-polymer hybrid nanoparticle:
system targeted to macrophages, Antimicrob. Agents Chemother. 42 (1998) biodistribution and toxicity assessment of encapsulated amphotericin B, Eur. J.
348–351. Pharm. Biopharm. 89 (2015) 62–73.
[91] D. Dasgupta, P. Chakraborty, M.K. Basu, Ligation of Fc receptor of macrophages [118] P.K. Gupta, A.K. Jaiswal, et al., Self-assembled ionically sodium alginate cross-
stimulates protein kinase C and anti-leishmanial activity, Mol. Cell. Biochem. 209 linked amphotericin B encapsulated glycol chitosan stearate nanoparticles:
(2000) 1–8. applicability in better chemotherapy and non-toxic delivery in visceral
[92] M. Mukherjee, W. Basu Ball, P.K. Das, Leishmania donovani activates SREBP2 to leishmaniasis, Pharm. Res. 32 (2015) 1727–1740.
modulate macrophage membrane cholesterol and mitochondrial oxidants for [119] R. Khatik, P. Dwivedi, et al., Development of targeted 1,2-diacyl-sn-glycero-3-
establishment of infection, Int. J. Biochem. Cell Biol. 55 (2014) 196–208. phospho-l-serine-coated gelatin nanoparticles loaded with amphotericin B for
[93] F. Frézard, C. Demicheli, New delivery strategies for the old pentavalent improved in vitro and in vivo effect in leishmaniasis, Expert Opin. Drug Deliv. 11
antimonial drugs, Expert Opin. Drug Deliv. 7 (12) (2010) 1343–1358. (2014) 633–646.
[94] A.J. Coukell, R.N. Brogden, Liposomal amphotericin B: therapeutic use in the [120] S. McClean, E. Prosser, E. Meehan, D. O’Malley, N. Clarke, Z. Ramtoola,
management of fungal infections and visceral leishmaniasis, Drugs 55 (4) (1998) D. Brayden, Binding and uptake of biodegradable poly-DL-lactide micro- and
585–612. nanoparticles in intestinal epithelia, Eur, J. Pharm. Sci. 6 (1998) 153–163.
[95] J. Téllez, et al., Use liposomal nanoformulations antileishmania therapy: [121] G. Mittal, D.K. Sahana, V. Bhardwaj, M.N.V. Ravi, Kumar, Estradiol loaded PLGA
challanges and perspective, J. Liposome Res. 31 (2) (2021) 169–176. nanoparticles for oral administration: effect of polymer molecular weight and
[96] S. Bhowmick, R. Ravindran, N. Ali, Gp63 in stable cationic liposomes confers copolymer composition on release behavior in vitro and in vivo, J. Control.
sustained vaccine immunity to susceptible BALB/c mice infected with Leishmania Release 119 (2007) 77–85.
donovani, Infect. Immun. 76 (3) (2008) 1003–1015. [122] H. Van De Ven, C. Paulussen, et al., PLGA nanoparticles and nanosuspensions
[97] M.A. Khan, M. Owais, Toxicity, stability and pharmacokinetics of amphotericin B with amphotericin B: potent in vitro and in vivo alternatives to Fungizone and
in immunomodulator tuftsin-bearing liposomes in a murine model, J. Antimicrob. AmBisome, J. Control. Release 161 (2012) 795–803.
Chemother. 58 (2006) 125–132. [123] R.F. de Carvalho, I.F. Ribeiro, et al., Leishmanicidal activity of amphotericin B
[98] A. Pardakhty, M. Shakibaie, H. Daneshvar, A. Khamesipour, T. Mohammadi- encapsulated in PLGA–DMSA nanoparticles to treat cutaneous leishmaniasis in
Khorsand, H. Forootanfar, Preparation and evaluation of niosomes containing C57BL/6 mice, Exp. Parasitol. 135 (2013) 217–222.
autoclaved Leishmania major: a preliminary study, J. Microencapsul. 29 (2012) [124] R. Kumar, G.C. Sahoo, K. Pandey, V. Das, P. Das, Study the effects of PLGA-PEG
219–224. encapsulated Amphotericin B nanoparticle drug delivery system against
[99] P. Khazaeli, I. Sharifi, E. Talebian, G. Heravi, E. Moazeni, M. Mostafavi, Anti- Leishmania donovani, Drug Deliv. 22 (2015) 383–388.
leishmanial effect of itraconazole niosome on in vitro susceptibility of Leishmania [125] M. Nahar, N.K. Jain, Preparation, characterization and evaluation of targeting
tropica, Environ. Toxicol. Pharmacol. 38 (2014) 205–211. potential of amphotericin b-loaded engineered PLGA nanoparticles, Pharm. Res.
[100] A.J. Baillie, G.H. Coombs, T.F. Dolan, J. Laurie, Non-ionic surfactant vesicles, 26 (2009) 2588–2598.
niosomes, as a delivery system for the anti-leishmanial drug, sodium [126] G. Shahnaz, B.J. Edagwa, J. McMillan, S. Akhtar, A. Raza, N.A. Qureshi,
stibogluconate, J. Pharm. Pharmacol. 38 (1986) 502–505. M. Yasinzai, H.E. Gendelman, Development of mannose-anchored thiolated

15
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

amphotericin B nanocarriers for treatment of visceral leishmaniasis, [154] D.P. Lage, P.A.F. Ribeiro, D.S. Dias, et al., Liposomal formulation of chimerat, a
Nanomedicine 12 (2017) 99–115. multiple t-cell epitope-containing recombinant protein, is a candidate vaccine for
[127] R.K. Das, V.L. Pachapur, L. Lonappan, M. Naghdi, R. Pulicharla, S. Maiti, human visceral leishmaniasis, Vaccines 8 (2020) 1–20.
M. Cledon, L.M.A. Dalila, S.J. Sarma, S.K. Brar, Biological synthesis of metallic [155] D.P. Lage, P.A.F. Ribeiro, D.S. Dias, et al., A candidate vaccine for human visceral
nanoparticles: plants, animals and microbial aspects, Nanotechnol. Environ. Eng. leishmaniasis based on a specific T cell epitope-containing chimeric protein
2 (2017) 1–21. protects mice against Leishmania infantum infection, npj Vaccin. 5 (2020).
[128] S. Das, P. Roy, S. Mondal, T. Bera, A. Mukherjee, One pot synthesis of gold [156] Y. Xia, Q. Fan, D. Hao, J. Wu, G. Ma, Z. Su, Chitosan-based mucosal adjuvants:
nanoparticles and application in chemotherapy of wild and resistant type visceral sunrise on the ocean, Vaccine 33 (2015) 5997–6010.
leishmaniasis, Colloids Surf. B Biointerfaces 107 (2013) 27–34. [157] M. Black, A. Trent, M. Tirrell, C. Olive, Advances in the design and delivery of
[129] M.A. Awad, E.M. Al Olayan, M.I. Siddiqui, N.M. Merghani, S.S.A. llahAlsaif, A. peptide subunit vaccines with a focus on toll-like receptor agonists, Expert Rev.
S. Aloufi, Antileishmanial effect of silver nanoparticles: Green synthesis, Vaccin. 9 (2010) 157–173.
characterization, in vivo and in vitro assessment, Biomed. Pharmacother. 137 [158] A.C. Rice-Ficht, A.M. Arenas-Gamboa, M.M. Kahl-McDonagh, T.A. Ficht,
(2021), 111294. Polymeric particles in vaccine delivery, Curr. Opin. Microbiol. 13 (2010)
[130] A. Ahmad, Y. Wei, S. Ullah, S.I. Shah, F. Nasir, A. Shah, Z. Iqbal, K. Tahir, U. 106–112.
A. Khan, Q. Yuan, Synthesis of phytochemicals-stabilized gold nanoparticles and [159] S.K. Mallapragada, B. Narasimhan, Immunomodulatory biomaterials, Int. J.
their biological activities against bacteria and Leishmania, Microb. Pathog. 110 Pharm. 364 (2008) 265–271.
(2017) 304–312. [160] K. Sehgal, K.M. Dhodapkar, M.V. Dhodapkar, targeting human dendritic cells in
[131] P. Kumar, P. Shivam, S. Mandal, et al., Synthesis, characterization, and situ to improve vaccines, Immunol. Lett. 162 (2014) 59–67.
mechanistic studies of a gold nanoparticle-amphotericin B covalent conjugate [161] K.D. Newman, J. Samuel, G. Kwon, Ovalbumin peptide encapsulated in poly (d,l
with enhanced antileishmanial efficacy and reduced cytotoxicity, Int. J. lactic-co-glycolic acid) microspheres is capable of inducing a T helper type 1
Nanomed. 14 (2019) 6073–6101. immune response, J. Control. Release 54 (1998) 49–59.
[132] D.K. Das, A. Chakraborty, S. Bhattacharjee, Biosynthesis of stabilised gold [162] M. Diwan, M. Tafaghodi, J. Samuel, Enhancement of immune responses by co-
nanoparticle using an aglycone flavonoid, quercetin, J. Exp. Nanosci. 8 (4) (2013) delivery of a CpG oligodeoxynucleotide and tetanus toxoid in biodegradable
649–655. nanospheres, J. Control. Release 85 (2002) 247–262.
[133] P. Prasanna, P. Kumar, S. Mandal, et al., 7,8–dihydroxyflavone functionalized [163] P.M.H. Heegaard, L. Dedieu, N. Johnson, M.F. Le Potier, M. Mockey, F. Mutinelli,
gold nanoparticles target the arginase enzyme of Leishmania donovani, Nanomed. T. Vahlenkamp, M. Vascellari, N.S. Sørensen, Adjuvants and delivery systems in
(Lond.) (2021), https://doi.org/10.2217/nnm-2021-0161. veterinary vaccinology: current state and future developments, Arch. Virol. 156
[134] A.M. Allahverdiyev, E.S. Abamor, et al., Antileishmanial effect of silver (2011) 183–202.
nanoparticles and their enhanced antiparasitic activity under ultraviolet light, Int. [164] D.T. O’Hogan, M. Singh, Microparticles as vaccine adjuvants and delivery
J. Nanomed. 6 (2011) 2705–2714. systems, Expert Rev. Vaccin. 2 (2003) 269–283.
[135] A. Ahmad, Y. Wei, F. Syed, S. Khan et al., Isatis tinctoria mediated synthesis of [165] N. Saljoughian, F. Zahedifard, D. Doroud, F. Doustdari, M. Vasei,
amphotericin B-bound silver nanoparticles with enhanced photoinduced B. Papadopoulou, S. Rafati, Cationic solid-lipid nanoparticles are as efficient as
antileishmanial activity: a novel green approach, J. Photochem. Photobiol. B Biol. electroporation in DNA vaccination against visceral leishmaniasis in mice,
161 (2016) 17–24. Parasite Immunol. 35 (2013) 397–408.
[136] A. Nadhman, M.I. Khan, S. Nazir, et al., Annihilation of Leishmania by daylight [166] I. Jafari, V. Heravi Shargh, M. Shahryari, A. Abbasi, M.R. Jaafari,
responsive ZnO nanoparticles: a temporal relationship of reactive oxygen species- A. Khamesipour, A. Badiee, Cationic liposomes formulated with a novel whole
induced lipid and protein oxidation, Int. J. Nanomed. 11 (2016) 2451–2461.7. Leishmania lysate (WLL) as a vaccine for leishmaniasis in murine model,
[137] S.K. Kalangi, A. Dayakar, D. Gangappa, et al., Biocompatible silver nanoparticles Immunobiology 223 (2018) 493–500.
reduced from Anethum graveolens leaf extract augments the antileishmanial [167] H. Firouzmand, M. Sahranavard, A. Badiee, A. Khamesipour, S.H. Alavizadeh,
efficacy of miltefosine, Exp. Parasitol. 170 (2016) 184–192. A. Samiei, D. Soroush, M. TavassotiKheiri, F. Mahboudi, M.R. Jaafari, The role of
[138] A. Ahmad, F. Syed, M. Imran, et al., Phytosynthesis and antileishmanial activity of LPD-nanoparticles containing recombinant major surface glycoprotein of
gold nanoparticles by M aytenus Royleanus, J. Food Biochem. 40 (2016) Leishmania (rgp63) in protection against leishmaniasis in murine model,
420–427. Immunopharmacol. Immunotoxicol. 40 (2018) 72–82.
[139] A.A. Zahir, I.S. Chauhan, A. Bagavan, C. Kamaraj, G. Elango, J. Shankar, [168] J.D. Ospina-Villa, C. Gómez-Hoyos, R. Zuluaga-Gallego, O. Triana-Chávez,
N. Arjaria, S.M. Roopan, A.A. Rahuman, N. Singh, Green synthesis of silver and Encapsulation of proteins from Leishmania panamensis into PLGA particles by a
titanium dioxide nanoparticles using Euphorbia prostrata extract shows shift from single emulsion-solvent evaporation method, J. Microbiol. Methods 162 (2019)
apoptosis to G0/G1 arrest followed by necrotic cell death in Leishmania 1–7.
donovani, Antimicrob. Agents Chemother. 59 (2015) 4782–4799. [169] S. Zutshi, et al., Anti-leishmanial vaccines: assumptions, approaches, and
[140] S. Ghosh, S. Jagtap, P. More, U.J. Shete, N.O. Maheshwari, S.J. Rao, R. Kitture, annulments, Vaccines 7 (4) (2019) 156.
S. Kale, J. Bellare, S. Patil, J.K. Pal, B.A. Chopade, Dioscorea bulbifera mediated [170] J.M. Silverman, et al., Leishmania Exosomes modulate innate and adaptive
synthesis of novel AucoreAgshell nanoparticles with potent antibiofilm and immune responses through effects on monocytes and dendritic cells, J. Immunol.
antileishmanial activity, J. Nanomater. (2015) (2015). 185 (9) (2010) 5011–5022.
[141] M. Khatami, H. Alijani, I. Sharifi, F. Sharifi, S. Pourseyedi, S. Kharazi, M.A. Lima [171] P. Baiocco, A. Ilari, P. Ceci, S. Orsini, M. Gramiccia, T. Di Muccio, G. Colotti,
Nobre, M. Khatami, Leishmanicidal activity of biogenic Fe3O4 nanoparticles, Sci. Inhibitory effect of silver nanoparticles on trypanothione reductase activity and
Pharm. 85 (2017) 36. Leishmania infantum proliferation, ACS Med. Chem. Lett. 2 (2011) 230–233.
[142] Sumaira, M.S. Afridi, S.S. Hashmi, G.S. Ali, M. Zia, B.H. Abbasi, Comparative [172] S. Hussain, S. Garantziotis, F. Rodrigues-Lima, J.M. Dupret, A. Baeza-Squiban,
antileishmanial efficacy of the biosynthesized ZnO NPs from genus Verbena, IET S. Boland, Intracellular signal modulation by nanomaterials, Adv. Exp. Med. Biol.
Nanobiotechnol. 12 (2018) 1067–1073. 811 (2014) 113–134.
[143] A.B. Djurišić, et al., Toxicity of metal oxide nanoparticles: mechanisms, [173] M.Y. Want, et al., Therapeutic efficacy of artemisinin-loaded nanoparticles in
characterization, and avoiding experimental artefacts, Small 11 (1) (2015) 26–44. experimental visceral leishmaniasis, Colloids Surf. B Biointerfaces 130 (2015)
[144] A.B. Sengul, E. Asmatulu, Toxicity of metal and metal oxide nanoparticles: a 215–221.
review, Environ. Chem. Lett. 18 (5) (2020) 1659–1683. [174] A. Roy, O. Bulut, S. Some, A.K. Mandal, M.D. Yilmaz, Green synthesis of silver
[145] Y.S. Chen, Y.C. Hung, I. Liau, G.S. Huang, Assessment of the in vivo toxicity of nanoparticles: biomolecule-nanoparticle organizations targeting antimicrobial
gold nanoparticles, Nanoscale Res. Lett. 4 (8) (2009) 858–864. activity, RSC Adv. 9 (5) (2019) 2673–2702.
[146] A. Balfourier, et al., Unexpected intracellular biodegradation and recrystallization [175] J.D.F. Inacio, M.M. Canto-Cavalheiro, R.F.S. Menna-Barreto, E.E. Almeida-
of gold nanoparticles, Proc. Natl. Acad. Sci. USA 117 (1) (2020) 103–113. Amaral, Mitochondrial damage contribute to epigallocatechin-3-gallate induced
[147] K. Saleem, Z. Khursheed, C. Hano, I. Anjum, S. Anjum, Applications of death in Leishmania amazonensis, Exp. Parasitol. 132 (2) (2012) 151–155.
nanomaterials in leishmaniasis: a focus on recent advances and challenges, [176] T. Pacheco-Fernandez, G. Volpedo, C. Verma, A.R. Satoskar, Understanding the
Nanomaterials 9 (12) (2019) 1749. immune responses involved in mediating protection or immunopathology during
[148] D. Doroud, S. Rafati, Leishmaniasis: focus on the design of nanoparticulate leishmaniasis, Biochem. Soc. Trans. 49 (2021) 297–311.
vaccine delivery systems, Expert Rev. Vaccin. 11 (2012) 69–86. [177] C.L. Ventola, Progress in nanomedicine: approved and investigational nanodrugs,
[149] E. Handman, Leishmaniasis: current status of vaccine development, Clin. P T 42 (2017) 742–755.
Microbiol. Rev. 14 (2001) 229–243. [178] J.M. Caster, A.N. Patel, T. Zhang, A. Wang, Investigational nanomedicines in
[150] M.S. Duthie, V.S. Raman, F.M. Piazza, S.G. Reed, The development and clinical 2016: a review of nanotherapeutics currently undergoing clinical trials, Wiley
evaluation of second-generation leishmaniasis vaccines, Vaccine 30 (2012) Interdiscip. Rev. Nanomed. Nanobiotechnol. 9 (1) (2017).
134–141. [179] J. Roychoudhury, R. Sinha, N. Ali, Therapy with sodium stibogluconate in
[151] E.M. Janssen, E.E. Lemmens, T. Wolfe, et al., CD4+ T cells are required for stearylamine-bearing liposomes confers cure against SSG-resistant leishmania
secondary expansion and memory in CD8+ T lymphocytes, Nature 421 (2003) donovani in BALB/c mice, PLoS One 6 (2011), e17376.
852–856. [180] N.R.H. Stone, T. Bicanic, R. Salim, W. Hope, Liposomal Amphotericin B
[152] P.M. Gillespie, C.M. Beaumier, U. Strych, et al., Status of vaccine research and (AmBisome®): a review of the pharmacokinetics, pharmacodynamics, clinical
development of vaccines for leishmaniasis, Vaccine 34 (2016) 2992–2995. experience and future directions, Drugs 76 (2016) 485–500.
[153] G. Sharma, S. Kar, W. Basu Ball, K. Ghosh, P.K. Das, The curative effect of [181] A.K. Agrawal, A. Agrawal, A. Pal, P.Y. Guru, C.M. Gupta, Superior
fucoidan on visceral leishmaniasis is mediated by activation of MAP kinases chemotherapeutic efficacy of Amphotericin B in tuftsin-bearing liposomes against
through specific protein kinase C isoforms, Cell. Mol. Immunol. 11 (2014) Leishmania donovani infection in hamsters, J. Drug Target. 10 (2002) 41–45.
263–274.

16
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

[182] L. Kole, L. Das, P.K. Das, Synergistic effect of interferon-γ and mannosylated [206] S. Espuelas, P. Legrand, P.M. Loiseau, C. Bories, G. Barratt, J.M. Irache, In vitro
liposome- incorporated doxorubicin in the therapy of experimental visceral reversion of amphotericin B resistance in Leishmania donovani by Poloxamer
leishmaniasis, J. Infect. Dis. 180 (1999) 811–820. 188, Antimicrob. Agents Chemother. 44 (2000) 2190–2192.
[183] M.Y. Want, M. Islammudin, G. Chouhan, H.A. Ozbak, H.A. Hemeg, A. [207] A.G. Tempone, D. Perez, S. Rath, A.L. Vilarinho, R.A. Mortara, H.F. de Andrade,
P. Chattopadhyay, F. Afrin, Nanoliposomal artemisinin for the treatment of Targeting Leishmania (L.) chagasi amastigotes through macrophage scavenger
murine visceral leishmaniasis, Int. J. Nanomed. 12 (2017) 2189–2204. receptors: the use of drugs entrapped in liposomes containing phosphatidylserine,
[184] P.K. Gupta, A.K. Jaiswal, V. Kumar, A. Verma, P. Dwivedi, A. Dube, P.R. Mishra, J. Antimicrob. Chemother. 54 (2004) 60–68.
Covalent functionalized self-assembled lipo-polymerosome bearing amphotericin [208] D.A. Goyeneche-Patino, L. Valderrama, J. Walker, N.G. Saravia, Antimony
B for better management of leishmaniasis and its toxicity evaluation, Mol. Pharm. resistance and trypanothione in experimentally selected and clinical strains of
11 (2014) 951–963. Leishmania panamensis, Antimicrob. Agents Chemother. 52 (2008) 4503–4506.
[185] J. Sinha, S. Mukhopadhyay, N. Das, M.K. Basu, Targeting of liposomal [209] S. Nicoletti, K. Seifert, I.H. Gilbert, N-(2-hydroxypropyl)methacrylamide-
andrographolide to L. donovani-infected macrophages in vivo, Drug Deliv. J. amphotericin B (HPMA-AmB) copolymer conjugates as antileishmanial agents,
Deliv. Target. Ther. Agents 7 (2000) 209–213. Int. J. Antimicrob. Agents 33 (2009) 441–448.
[186] J. Ghosh, R. Guha, S. Das, S. Roy, Liposomal cholesterol delivery activates the [210] V.K. Prajapati, K. Awasthi, S. Gautam, T.P. Yadav, M. Rai, O.N. Srivastava,
macrophage innate immune arm to facilitate intracellular leishmania donovani S. Sundar, Targeted killing of leishmania donovani in vivo and in vitro with
killing, Infect. Immun. 82 (2014) 607–617. amphotericin β attached to functionalized carbon nanotubes, J. Antimicrob.
[187] E. Peretti, I. Miletto, B. Stella, F. Rocco, G. Berlier, S. Arpicco, Strategies to obtain Chemother. 66 (2011) 874–879.
encapsulation and controlled release of pentamidine in mesoporous silica [211] S.L. Mudavath, M. Talat, M. Rai, O.N. Srivastava, S. Sundar, Characterization and
nanoparticles, Pharmaceutics 10 (2018) 195. evaluation of amine-modified graphene amphotericin B for the treatment of
[188] T.G. Ribeiro, M.A. Chávez-Fumagall, D.G. Valadares, J.R. França, L.B. Rodrigues, visceral leishmaniasis: In vivo and in vitro studies, Drug Des. Devel. Ther. 8
M.C. Duarte, P.S. Lage, P.H.R. Andrade, D.P. Lage, L.V. Arruda, C. Refidaff, L. (2014) 1235–1247.
E. Costa, V.T. Martins, C.A.P. Tavares, R.O. Castilho, E.A.F. Coelho, A.A. [212] S. Kansal, R. Tandon, P. Dwivedi, P. Misra, P.R.P. Verma, A. Dube, P.R. Mishra,
G. Faraco, Novel targeting using nanoparticles: an approach to the development Development of nanocapsules bearing doxorubicin for macrophage targeting
of an effective anti-leishmanial drug-delivery system, Int. J. Nanomed. 9 (2014) through the phosphatidylserine ligand: a system for intervention in visceral
877–890. leishmaniasis, J. Antimicrob. Chemother. 67 (2012) 2650–2660.
[189] D. Barros, S.A.C. Lima, A. Cordeiro-Da-Silva, Surface functionalization of [213] V.K. Prajapati, K. Awasthi, T.P. Yadav, M. Rai, O.N. Srivastava, S. Sundar, An oral
polymeric nanospheres modulates macrophage activation: relevance in formulation of amphotericin B attached to functionalized carbon nanotubes is an
Leishmaniasis therapy, Nanomedicine 10 (2015) 387–403. effective treatment for experimental visceral leishmaniasis, J. Infect. Dis. 205
[190] S.A. Costa Lima, R. Silvestre, D. Barros, J. Cunha, M.T. Baltazar, R.J. Dinis- (2012) 333–336.
Oliveira, A. Cordeiro-da-Silva, Crucial CD8+ T-lymphocyte cytotoxic role in [214] S. Asthana, P.K. Gupta, A.K. Jaiswal, A. Dube, M.K. Chourasia, Targeted
amphotericin B nanospheres efficacy against experimental visceral leishmaniasis, chemotherapy of visceral leishmaniasis by lactoferrin-appended amphotericin B-
Nonomedicine 10 (2014) e1021–e1030. loaded nanoreservoir: In vitro and in vivo studies, Nanomedicine 10 (2015)
[191] J.A. Sánchez-Brunete, M.A. Dea, S. Rama, F. Bolás, J.M. Alunda, R. Raposo, M. 1093–1109.
T. Méndez, S. Torrado-Santiago, J.J. Torrado, Treatment of experimental visceral [215] S. Kunjachan, S. Gupta, A.K. Dwivedi, A. Dube, M.K. Chourasia, Chitosan-based
leishmaniasis with amphotericin B in stable albumin microspheres, Antimicrob. macrophage-mediated drug targeting for the treatment of experimental visceral
Agents Chemother. 48 (2004) 3246–3252. leishmaniasis, J. Microencapsul. 28 (2011) 301–310.
[192] M. Chaurasia, V.K. Pawar, A.K. Jaiswal, A. Dube, S. k Paliwal, M.K. Chourasia, [216] D.M. Casa, D.B. Scariot, N.M. Khalil, C.V. Nakamura, R.M. Mainardes, Bovine
Chondroitin nanocapsules enhanced doxorubicin induced apoptosis against serum albumin nanoparticles containing amphotericin B were effective in treating
leishmaniasis via Th1 immune response, Int. J. Biol. Macromol. 79 (2015) 27–36. murine cutaneous leishmaniasis and reduced the drug toxicity, Exp. Parasitol. 192
[193] S. Kansal, R. Tandon, P.R.P. Verma, A. Dube, P.R. Mishra, Development of (2018) 12–18.
doxorubicin loaded novel core shell structured nanocapsules for the intervention [217] A. Barazesh, M.H. Motazedian, N. Sattarahmady, M.H. Morowvat, S. Rashidi,
of visceral leishmaniasis, J. Microencapsul. 30 (2013) 441–450. Preparation of meglumine antimonate loaded albumin nanoparticles and
[194] P. Chaubey, B. Mishra, Mannose-conjugated chitosan nanoparticles loaded with evaluation of its anti-leishmanial activity: an in vitro assay, J. Parasit. Dis. 42
rifampicin for the treatment of visceral leishmaniasis, Carbohydr. Polym. 101 (2018) 416–422.
(2014) 1101–1108. [218] M. Nahar, V. Dubey, D. Mishra, P.K. Mishra, A. Dube, N.K. Jain, In vitro
[195] M.R. Gedda, P. Madhukar, A.K. Vishwakarma, V. Verma, A.K. Kushwaha, evaluation of surface functionalized gelatin nanoparticles for macrophage
G. Yadagiri, S.L. Mudavath, O.P. Singh, O.N. Srivastava, S. Sundar, Evaluation of targeting in the therapy of visceral leishmaniasis, J. Drug Target. 18 (2010)
safety and antileishmanial efficacy of amine functionalized carbon-based 93–105.
composite nanoparticle appended with amphotericin B: an in vitro and preclinical [219] R.X. Armijos, M.M. Weigel, M. Calvopina, A. Hidalgo, W. Cevallos, J. Correa,
study, Front. Chem. 8 (2020). Safety, immunogenecity, and efficacy of an autoclaved Leishmania amazonensis
[196] J.L. Italia, M.N.V. Ravi Kumar, K.C. Carter, Evaluating the potential of polyester vaccine plus BCG adjuvant against New World cutaneous leishmaniasis, Vaccine
nanoparticles for per oral delivery of amphotericin B in treating visceral 22 (2004) 1320–1326.
leishmaniasis, J. Biomed. Nanotechnol. 8 (2012) 695–702. [220] E.A.G. Khalil, A.M. Musa, F. Modabber, A.M. El-Hassan, Safety and
[197] P. Saudagar, V.K. Dubey, Carbon nanotube based betulin formulation shows immunogenicity of a candidate vaccine for visceral leishmaniasis (Alum-
better efficacy against Leishmania parasite, Parasitol. Int. 63 (2014) 772–776. precipitated autoclaved Leishmania major + BCG) in children: an extended phase
[198] S. Ghosh, N. Kar, T. Bera, Oleanolic acid loaded poly lactic co- glycolic acid- II study, Ann. Trop. Paediatr. 26 (2006) 357–361.
vitamin E TPGS nanoparticles for the treatment of Leishmania donovani infected [221] I.N. Satti, H.Y. Osman, N.S. Daifalla, S.A. Younis, E.A.G. Khalil, E.E. Zijlstra, A.
visceral leishmaniasis, Int. J. Biol. Macromol. 93 (2016) 961–970. M. El Hassan, H.W. Ghalib, Immunogenicity and safety of autoclaved Leishmania
[199] P. Roy, S. Das, T. Bera, S. Mondol, A. Mukherjee, Andrographolide nanoparticles major plus BCG vaccine in healthy Sudanese volunteers, Vaccine 19 (2001)
in leishmaniasis: characterization and in vitro evaluations, Int. J. Nanomed. 5 2100–2106.
(2010) 1113–1121. [222] G.P. Borja-Cabrera, N.N. Correia Pontes, V.O. Da Silva, E. Paraguai De Souza, W.
[200] P.E. Feuser, M.L. Tonini, A.V. Jacques, M.C. Santos da Silva, M. Steindel, C. Sayer, R. Santos, E.M. Gomes, K.G. Luz, M. Palatnik, C.B. Palatnik, De Sousa, Long
P.H. Hermes, de Araújo, Increased in vitro leishmanicidal activity of octyl gallate lasting protection against canine kala-azar using the FML-QuilA saponin vaccine
loaded poly(methyl methacrylate) nanoparticles, Pharm. Dev. Technol. 24 (2019) in an endemic area of Brazil (São Gonçalo do Amarante, RN), Vaccine 20 (2002)
593–599. 3277–3284.
[201] R. Gaspar, F.R. Opperdoes, V. Preat, M. Roland, Drug targeting with [223] V. Martin, I. Vouldoukis, J. Moreno, D. McGahie, S. Gueguen, A.M. Cuisinier, The
polyalkylcyanoacrylate nanoparticles: In vitro activity of primaquine-loaded protective immune response produced in dogs after primary vaccination with the
nanoparticles against intracellular Leishmania donovani, Ann. Trop. Med. LiESP/QA-21 vaccine (CaniLeish®) remains effective against an experimental
Parasitol. 86 (1992) 41–49. challenge one year later, Vet. Res. 45 (2014).
[202] C. de C. Spadari, F.W.M. da S. de Bastiani, L.B. Lopes, K. Ishida, <p>Alginate [224] S. Datta, S. Roy, M. Manna, Therapy with radio-attenuated vaccine in
nanoparticles as non-toxic delivery system for miltefosine in the treatment of experimental murine visceral leishmaniasis showed enhanced T cell and inducible
candidiasis and cryptococcosis<p>, Int. J. Nanomedicine. Volume 14 (2019) nitric oxide synthase levels, suppressed tumor growth factor-beta production with
5187–5199. higher expression of some signaling molecules, Brazilian, J. Infect. Dis. 19 (2015)
[203] S. Mondal, P. Roy, S. Das, A. Halder, A. Mukherjee, T. Bera, In Vitro 36–42.
susceptibilities of wild and drug resistant Leishmania donovani amastigote stages [225] H. Daneshvar, G.H. Coombs, P. Hagan, R.S. Phillips, Leishmania mexicana and
to andrographolide nanoparticle: role of vitamin e derivative TPGS for Leishmania major: attenuation of wild-type parasites and vaccination with the
nanoparticle efficacy, PLoS One 8 (2013). attenuated lines, J. Infect. Dis. 187 (2003) 1662–1668.
[204] A.H.A. Mohamed-Ahmed, K. Seifert, V. Yardley, H. Burrell-Saward, S. Brocchini, [226] R.N. Coler, M.S. Duthie, K.A. Hofmeyer, J. Guderian, L. Jayashankar, J. Vergara,
S.L. Croft, Antileishmanial activity, uptake, and biodistribution of an T. Rolf, A. Misquith, J.D. Laurance, V.S. Raman, H.R. Bailor, N.D. Cauwelaert, S.
amphotericin B and poly(α-glutamic acid) complex, Antimicrob. Agents J. Reed, A. Vallur, M. Favila, M.T. Orr, J. Ashman, P. Ghosh, D. Mondal, S.
Chemother. 57 (2013) 4608–4614. G. Reed, From mouse to man: safety, immunogenicity and efficacy of a candidate
[205] A.D. Duong, S. Sharma, K.J. Peine, G. Gupta, A.R. Satoskar, E.M. Bachelder, B. leishmaniasis vaccine LEISH-F3+GLA-SE, Clin. Transl. Immunol. 4 (2015).
E. Wyslouzil, K.M. Ainslie, Electrospray encapsulation of toll-like receptor agonist [227] G. Grimaldi, A. Teva, C.B. Dos-Santos, F.N. Santos, I.D.S. Pinto, B. Fux, G.R. Leite,
resiquimod in polymer microparticles for the treatment of visceral leishmaniasis, A. Falqueto, Field trial of efficacy of the Leish-tec® vaccine against canine
Mol. Pharm. 10 (2013) 1045–1055. leishmaniasis caused by Leishmania infantum in an endemic area with high
transmission rates, PLoS One 12 (2017).

17
P. Prasanna et al. Biomedicine & Pharmacotherapy 141 (2021) 111920

[228] M. Moafi, R. Sherkat, R. Taleban, H. Rezvan, Leishmania vaccines entered in [242] A. Das, N. Ali, Combining cationic liposomal delivery with MPL-TDM for cysteine
clinical trials: a review of literature, Int. J. Prev. Med. 10 (2019) 1–6. protease cocktail vaccination against leishmania donovani: evidence for antigen
[229] M. Agallou, M. Margaroni, E. Karagouni, Cellular vaccination with bone marrow- synergy and protection, PLOS Trop. Dis. 8 (2014), e3091.
derived dendritic cells pulsed with a peptide of Leishmania infantum KMP-11 and [243] F. Eskandari, G.A. Talesh, M. Parooie, M.R. Jaafari, A. Khamesipour, Z. Saberi,
CpG oligonucleotides induces protection in a murine model of visceral A. Abbasi, A. Badiee, Immunoliposomes containing Soluble Leishmania Antigens
leishmaniasis, Vaccine 29 (2011) 5053–5064. (SLA) as a novel antigen delivery system in murine model of leishmaniasis, Exp.
[230] R. Choudhury, P. Das, T. De, T. Chakraborti, 115 kDa serine protease confers Parasitol. 146 (2014) 78–86.
sustained protection to visceral leishmaniasis caused by Leishmania donovani via [244] A.P.V. Moura, L.C.B. Santos, C.R.N. Brito, E. Valencia, C. Junqueira, A.A.P. Filho,
IFN-γ induced down-regulation of TNF-α mediated MMP-9 activity, M.R.V. Sant’Anna, N.F. Gontijo, D.C. Bartholomeu, R.T. Fujiwara, R.T. Gazzinelli,
Immunobiology 218 (2013) 114–126. C.S. McKay, C.A. Sanhueza, M.G. Finn, A.F. Marques, Virus-like particle display of
[231] M.E.E. Elfaki, E.A.G. Khalil, A.S. De Groot, A.M. Musa, A. Gutierrez, B.M. Younis, the α-gal carbohydrate for vaccination against leishmania infection, ACS Cent.
K.A.M. Salih, A.M. El-Hassan, Immunogenicity and immune modulatory effects of Sci. 3 (2017) 1026–1031.
in silico predicted L. donovani candidate peptide vaccines, Hum. Vaccin. [245] A. Katebi, R. Varshochian, F. Riazi-rad, M. Ganjalikhani-Hakemi, S. Ajdary,
Immunother. 8 (2012) 1769–1774. Combinatorial delivery of antigen and TLR agonists via PLGA nanoparticles
[232] M. Osman, A. Mistry, A. Keding, R. Gabe, E. Cook, S. Forrester, R. Wiggins, S. Di modulates Leishmania major-infected-macrophages activation, Biomed.
Marco, S. Colloca, L. Siani, D.F. Smith, T. Aebischer, P.M. Kaye, C.J. Lacey, Pharmacother. 137 (2021), 111276.
A third generation vaccine for human visceral leishmaniasis and post kala azar [246] O.A. Tosyali, A. Allahverdiyev, M. Bagirova, E.S. Abamor, M. Aydogdu,
dermal leishmaniasis: first-in-human trial of ChAd63-KH, Plos. Trop. Dis. 11 S. Dinparvar, T. Acar, Z. Mustafaeva, S. Derman, Nano-co-delivery of
(2017). lipophosphoglycan with soluble and autoclaved leishmania antigens into PLGA
[233] A. Mehravaran, M.R. Nasab, H. Mirahmadi, I. Sharifi, E. Alijani, A.R. Nikpoor, nanoparticles: evaluation of in vitro and in vivo immunostimulatory effects
J. Akhtari, Protection induced by Leishmania Major antigens and the imiquimod against visceral leishmaniasis, Mater. Sci. Eng. C 120 (2021), 111684.
adjuvant encapsulated on liposomes in experimental cutaneous leishmaniasis, [247] H. Noormehr, A. Zavaran Hosseini, S. Soudi, F. Beyzay, Enhancement of Th1
Infect. Genet. Evol. 70 (2019) 27–35. immune response against Leishmania cysteine peptidase A, B by PLGA
[234] R. Ravindran, M. Maji, N. Ali, Vaccination with liposomal leishmanial antigens nanoparticle, Int. Immunopharmacol. 59 (2018) 97–105.
adjuvanted with monophosphoryl lipid-trehalose dicorynomycolate (MPL-TDM) [248] E. Athanasiou, M. Agallou, S. Tastsoglou, O. Kammona, A. Hatzigeorgiou,
confers long-term protection against visceral leishmaniasis through a human C. Kiparissides, E. Karagouni, A poly(lactic-co-glycolic) acid nanovaccine based
administrable route, Mol. Pharm. 9 (2012) 59–70. on chimeric peptides from different Leishmania infantum proteins induces
[235] A. Badiee, M.R. Jaafari, A. Khamesipour, A. Samiei, D. Soroush, M.T. Kheiri, dendritic cells maturation and promotes peptide-specific IFNγ-producing CD8+ T
F. Barkhordari, W.R. McMaster, F. Mahboudi, The role of liposome charge on cells essential for the protection against experiment, Front. Immunol. 8 (2017)
immune response generated in BALB/c mice immunized with recombinant major 684.
surface glycoprotein of Leishmania (rgp63), Exp. Parasitol. 121 (2009) 362–369. [249] M. Hojatizade, M. Soleymani, M. Tafaghodi, A. Badiee, O. Chavoshian, M.
[236] S. Mazumder, R. Ravindran, A. Banerjee, N. Ali, Non-coding pDNA bearing R. Jaafari, Chitosan nanoparticles loaded with whole and soluble leishmania
immunostimulatory sequences co-entrapped with leishmanial antigens in cationic antigens, and evaluation of their immunogenecity in a mouse model of
liposomes elicits almost complete protection against experimental visceral leishmaniasis, Iran. J. Immunol. 15 (2018) 281–293.
leishmaniasis in BALB/c mice, Vaccine 25 (2007) 8771–8781. [250] S. Zarrati, M. Mahdavi, F. Tabatabaie, Immune responses in DNA vaccine
[237] V. Heravi Shargh, M.R. Jaafari, A. Khamesipour, S.A. Jalali, H. Firouzmand, formulated with PMMA following immunization and after challenge with
A. Abbasi, A. Badiee, Cationic liposomes containing soluble Leishmania antigens Leishmania major, J. Parasit. Dis. 40 (2016) 427–435.
(SLA) plus CpG ODNs induce protection against murine model of leishmaniasis, [251] K.G. Nelson, J.V. Bishop, R.O. Ryan, R. Titus, Nanodisk-associated amphotericin B
Parasitol. Res. 111 (2012) 105–114. clears Leishmania major cutaneous infection in susceptible BALB/c mice,
[238] P.A.F. Ribeiro, D.S. Dias, M.V.M. Novais, D.P. Lage, G.S.V. Tavares, D.V. Antimicrob. Agents Chemother. 50 (2006) 1238–1244.
C. Mendonça, J.S. Oliveira, M.A. Chávez-Fumagalli, B.M. Roatt, M.C. Duarte, [252] M. Ghadiri, A. Vatanara, D. Doroud, A.R. Najafabadi, Paromomycin loaded solid
D. Menezes-Souza, F. Ludolf, C.A.P. Tavares, M.C. Oliveira, E.A.F. Coelho, lipid nanoparticles: characterization of production parameters, Biotechnol.
A. Leishmania, hypothetical protein-containing liposome-based formulation is Bioprocess Eng. 16 (2011) 617–623.
highly immunogenic and induces protection against visceral leishmaniasis, [253] M.A. Danesh-Bahreini, J. Shokri, A. Samiei, E. Kamali-Sarvestani, M. Barzegar-
Cytokine 111 (2018) 131–139. Jalali, S. Mohammadi-Samani, Nanovaccine for leishmaniasis: preparation of
[239] M. Hojatizade, A. Badiee, A. Khamesipour, A. Mirshafiey, J. Akhtari, chitosan nanoparticles containing Leishmania superoxide dismutase and
A. Mehravaran, S. HodaAlavizadeh, A. Abbasi, Z. Saberi, A.R. Nikpoor, M. evaluation of its immunogenicity in BALB/c mice, Int. J. Nanomed. 6 (2011)
R. Jaafari, DDA/TDB liposomes containing soluble Leishmania major antigens 835–842.
induced a mixed Th1/Th2 immune response in BALB/c mice, Nanomed. J. 4 [254] S.K. Sharma, A. Dube, A. Nadeem, S. Khan, I. Saleem, R. Garg, O. Mohammad,
(2017) 71–82. Non PC liposome entrapped promastigote antigens elicit parasite specific CD8+
[240] F. Fakhraee, A. Badiee, S.H. Alavizadeh, S.A. Jalali, O. Chavoshian, and CD4+ T-cell immune response and protect hamsters against visceral
A. Khamesipour, F. Mahboudi, M.R. Jaafari, Coadminstration of L. major leishmaniasis, Vaccine 24 (2006) 1800–1810.
amastigote class I nuclease (rLmaCIN) with LPD nanoparticles delays the [255] D. Alti, M.V. Rao, D.N. Rao, R. Maurya, S.K. Kalangi, Gold–silver bimetallic
progression of skin lesion and the L. major dissemination to the spleen in BALB/c nanoparticles reduced with herbal leaf extracts induce ROS-mediated death in
mice-based experimental setting, Acta Trop. 159 (2016) 211–218. both promastigote and amastigote stages of leishmania donovani, ACS Omega 5
[241] A. Thakur, H. Kaur, S. Kaur, Studies on the protective efficacy of freeze thawed (2020) 16238–16245.
promastigote antigen of Leishmania donovani along with various adjuvants [256] S. Das, A. Halder, S. Mandal, M.A.J. Mazumder, T. Bera, A. Mukherjee, P. Roy,
against visceral leishmaniasis infection in mice, Immunobiology 220 (2015) Andrographolide engineered gold nanoparticle to overcome drug resistant
1031–1038. visceral leishmaniasis, Artif. Cells Nanomed. Biotechnol. 46 (2018) 751–762.

18

You might also like