Bacopasaponin C Critical Evaluation of Anti-Leishmanial Properties in Various Delivery Modes

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Drug Delivery

ISSN: 1071-7544 (Print) 1521-0464 (Online) Journal homepage: https://www.tandfonline.com/loi/idrd20

Bacopasaponin C: Critical Evaluation of Anti-


Leishmanial Properties in Various Delivery Modes

J. Sinha, B. Raay, N. Das, S. Medda, S. Garai, S. B. Mahato & M. K. Basu

To cite this article: J. Sinha, B. Raay, N. Das, S. Medda, S. Garai, S. B. Mahato & M. K. Basu
(2002) Bacopasaponin C: Critical Evaluation of Anti-Leishmanial Properties in Various Delivery
Modes, Drug Delivery, 9:1, 55-62, DOI: 10.1080/107175402753413181

To link to this article: https://doi.org/10.1080/107175402753413181

Published online: 29 Sep 2008.

Submit your article to this journal

Article views: 154

View related articles

Citing articles: 5 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=idrd20
Drug Delivery, 9:55– 62, 2002
Copyright °c 2002 Taylor & Francis
1071-7544 /02 $12.00 + .00

Bacopasaponin C: Critical Evaluation of


Anti-Leishmanial Properties in Various Delivery Modes
J. Sinha, B. Raay, N. Das, S. Medda, S. Garai, S. B. Mahato, and M. K. Basu
Biomembrane Division, Indian Institute of Chemical Biology, Calcutta, India

therapeutic level of drug in the systemic circulation or at the spe-


Bacopasaponin C, an indigenous glycoside, was isolated from ciŽ c target organ site furthering a reduction in any undesirable
Indian medicinal plant Bacopa monniera (b. brahmi) and was tested side effects of the drug.
for antileishmanial properties both in free and in various delivery Among the different strategies for site-speciŽ c drug delivery,
modes, e.g., niosomes, microspheres, and nanoparticles that are
used now as alternatives to more commonly used liposomes. The colloidal/vesicular carriers were found to be effective because of
different vesicles were prepared by published protocols. The per- their structure function diversity in in vivo and in vitro milieus.
cent intercalation of Bacopasaponin C in liposomes, niosomes, and Colloidal carriers are of various types, such as liposomes, nio-
micropspheres determined at its absorption maximal (¸max = somes, nanoparticles, and microspheres. The most popular and
238 nm, 2 = 8:6 £ 103 M¡ 1 cm¡ 1 ) was found to be 30; for nanopar- well-studied colloidal drug delivery systems are the liposomes,
ticles it was 50. At equivalent dose of 1.75 mg/kg body weight, every
third day for a total of 6 doses in 15 days, Bacopasaponin C in all though they have some fundamental problems, such as sensi-
the vesicular forms was found to be very active. An inverse lin- tivity lamellae and difŽ culty in scaled up production (Guerrero
ear relationship between the efŽ cacy and the size of the vesicles et al. 1998). Thus, to avoid such limitations of liposomes, al-
was established. As analyzed from tissue histology, blood pathol- ternative drug delivery systems as niosomes, nanocapsules, and
ogy, and speciŽ c tests related to normal liver and kidney functions, micropheres are researched. Niosomes (Baillie et al. 1985) are
Bacopasaponi n C in each of the four vesicular forms was found to
be without any side effects. Thus, because of its indigenous origin made from nonionic surfactants, that are quite stable, biodegrad-
and non-toxic nature, Bacopasaponin C could very well be consid- able, and cheap. Polymeric delivery vesicles include the mi-
ered for application in the clinic through these alternative delivery crospheres and nanoparticles and are made up of cost-effective
modes. biodegradable polymers such as poly(esers), poly (orthoesters),
polyanhydrides, and different natural polymers. These vesicles
Keywords Bacapasaponin C, EfŽ cacy versus Size, Liposomes, are found to be highly stable and biocompatible (Anderson and
Microspheres, Nanocapsules, Niosomes Shive 1997; Guterres et al. 1995). These vesicles are also ca-
pable of encapsulating a wide variety of drugs and molecules
(Hiremath and Hota 1999; Kreuter 1996).
Biomedical science has illustrated the need to control, regu-
late, and target the release of drug in the body to the particular Our investigation aims to compare the structure and function
of four drug delivery vesicles—liposomes, niosomes, micro-
tissue or cells of interest. In general, a primary requirement has
spheres, and nanocapsules—on the basis of their size, efŽ cacy,
been to provide less frequent drug administration with a constant
and toxicity with liposomes as control. The efŽ cacy of these
vesicles is studied by incorporating an antileishmanial com-
pound, Bacopasaponin-C, extracted from an Indian medicinal
Received 15 May 2001; accepted 6 August 2001. plant Bacopa monniera (b. brahmi) against experimental leish-
Financial support from University Grants Commission, New Delhi,
India, and Dept. of Biotechnology, New Delhi, India, are gratefully maniasis in a hamster model. Bacopasaponin-C is a glycosidic
acknowledged. Thanks are also due to CSIR, New Delhi, India, for pseudojujobogenin (Garai et al. 1996) having two carbohydrate
providing Ž nancial assistance to M. K. Basu in the form of ES Scheme. residues, glucose and rhamnose, attached to the triterpenoid
We are extremely grateful for helpful suggestions of Prof. S. P. Moulik, aglycone part. The unique presence of a glucose residue equipped
Jadavpur University, for dynamic light scaterring measurements and the compound to be a self-targeting molecule that can be directed
Dr. S. Chakraborty, Calcutta University, for hisopathological studies.
Address correspondence to M. K. Basu, Indian Institute of Chemical toward the glucose receptor present on the macrophage surface.
Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta-700032, India. This facilitates a receptor-mediated drug delivery to the reticu-
E-mail: mukulkbasu@yahoo.com loendothelial system.

55
56 J. SINHA ET AL.

MATERIALS AND METHODS The percent intercalation of Bacopasaponin-C in various ves-


Polylactide-co-glycolide (PLGA) (Avr. Mol. wt. 113, 200); icular forms was calculated from a small aliquot taken and sol-
polylactide(PLA) (Avr. Mol. wt. 106000); gelatin; Span-40 ubilized in the required solvent, knowing ¸max (238 nm) and
phosphatidylethanolamine (PE); cholesterol; dicetyl phosphate 2M (8.6 £ 103 M¡1 cm¡1 ). For liposomes, niosomes, and
(DCP); Tween-80; Span-40; isopropylmyristate; fetal calf serum nanocapsules, the percent intercalation was found to be »30;
(FCS); and RPMI-1640 were purchased from Sigma Chemicals for microspheres, the value increased to 50.
(St. Louis, MO, USA). Bacopasaponin-C was supplied by our
Medicinal Chemistry Department (IICB) with optimum purity
and quality. All other chemicals were of analytical grade. Animal Experiments
A colony of Golden hamsters (Mesocristatus auratus) orig-
inally from Haffkine Research Institute, Bombay, was used to
maintain Leishmania donovani, strain AG83, from an Indian
Preparation of Different Vesicles Systems
Kala-azar patient by intracardial passage every 6 weeks. Each
Bacopasaponin-C – loaded micropheres were prepared by the
animal was infected through intracardial passage with 2 £ 106
solvent evaporation technique. In short, PLGA and Bacopasa-
amastigotes. A group of 18 hamsters of average body weight
ponin-C at ratio of 15:1 w/w was added to 2 ml of dichlorome-
100 gm was infected and ready for drug testing after 1 month.
thane. This was then added drop-wise into 100 ml of PBS
The hamsters were distributed into free drug, liposomal drug,
(pH 7.2) containing 1% gelatin as a dispersing agent. The solu-
niosomal drug, microspehuralated drug, nanocapsulated drug,
tion was emulsiŽ ed by stirring for 2 hr resulting in the evapora-
and untreated control groups.
tion of dichloromethane. The gelatin solution was centrifuged
For MTD determination, a single dose treatment was fol-
at 8K – 10K rpm. The pellet was washed twice with PBS, then
lowed using varying amounts such as 1 mg to 10 mg/kg-body
collected and re-suspended in PBS (Wang et al. 1996).
weight of free drug; 4 such doses were given through subcuta-
MLV liposomes were prepared using PE, cholesterol, and
neous route at an interval of 3 days. The MTD of the free drug
DCP in a molar ratio of 7:4:1. Bacopasaponin-C was added to
was found to be 1.75 mg/kg body weight (Figure 1).
chloroform-methanol (2:1 v/v) solution of the lipids in a ratio of
The chemotherapy of multiple dose treatment was followed,
1.16::10 w/w (Gregoriadis and Ryman 1972). A thin dry Ž lm of
with average body weight of hamster at »100 gm. In practice,
the lipids and the drug was made on the inner surface of a round
0.175 mg of Bacopasaponin-C encapsulated in 0.5 ml of liposos-
bottom  ask by evaporating the organic solvents in a rotating
mal, niosomal, microsphere, and nanocapsule suspension was
 ask evaporator. The suspension was sonicated for 30 sec at
injected subcutaneously in each hamster every 3 days for a total
4± C and unencapsulated Bacopasaponin-C was separated from
of 6 days in 15 days. Free Bacopasaponin-C at same dosage
the vesicle-trapped components by centrifugation at 105,000 g
(0.175 mg/0.5 ml PBS) was administered to one group. Para-
for 1 hr; the pellet was suspended in PBS.
site burden of spleen was assessed from stained smears using
Niosomes were prepared from the nonionic surfactant, sorbi-
Stauber’s formula (Stauber et al. 1958).
tan monopalmitate (Span-40), cholesterol, and DCP in the molar
ratio 1:0.5:1. Chloroform was added to Span 40 and cholesterol.
The mixture was heated to 60± C in a water bath to form a so- Total number of amastigotes
lution. After cooling, 1 mg of Bacopasaponin-C (per 10 mg of D (Number of amastigotes=hostcell nucleus)
Span 40) dissolved in 100 ¹l of DMSO was added and a thin
£(weight of spleen in mg) £ 2 £ 105
Ž lm prepared. The Ž lm was dried in an atmosphere of N2 and
kept overnight in desiccators. The Ž lm was swelled with 1 ml of
PBS (pH 7.2) for 1 hr and then sonicated for 30 sec. The entire Investigation of Drug Toxicity
suspension was centrifuged at 100,000 £ g for 40 min; the pellet SpeciŽ c serum enzyme levels and splenic tissue histopathol-
formed was suspended in PBS to form the niosomal suspension ogy were assessed to Ž nd out drug toxicity. Serum gluta-
(Yoshioka et al. 1994). mate, pyruvate transaminase, and serum alkaline phosphatase
Nanocapsules are prepared according to the method of Seyler (Reitman and Frenkel 1957; Kind and King 1954) were assayed
et al. (1995) with some modiŽ cations. In brief, polylactide to analyze the hepatotoxicity. Similarly serum urea and creati-
(70 mg) was dissolved in 10 ml of dichloromethane. Phospha- nine levels were measured to Ž nd nephrotoxicity of the drug. The
tidylethanolamine (100 mg) was dissolved in 250 ¹l of isopropy- splenic histopathology was examined through haematoxylin-
lmyristate along with drug (9 mg) and added to dichloromethane eosin stained splenic section (Gurr 1962).
phase. The organic solution was added or allowed to run slowly
into 50 ml of PBS (pH 7.2) containing 0.4% of nonionic surfac-
tant (Tween-80), under moderate magnetic stirring. The organic Determination of Vesicular Size
phase was evaporated out by moderate stirring for 3 hr. This solu- The average vesicle diameters of the niosomes and nanocap-
tion was then ultracentrifuged at 35 K rpm, and the nanocapsules sules were determined through a dynamic light scattering
were then collected (Seyler et al. 1995). instrument (Otsuka Electronics, Japan) using Neon laser of
BACOPASAPONIN C 57

FIG. 1. MTD determination curve of Bacopasaponin-C , assessed through single dose treatment using varying amounts of 1 – 10 mg/kg body weight. Four such
doses were given through subcutaneou s route at intervals of 3 days.

¸ D 632:2 nm and taking measurements at 90± angle (Majhi RESULTS


and Moulik 1999; Moulik et al. 1999). The same for micro-
Size Determination and Distribution
spheres and liposomes was carried out with light microscopy to
get optimum and accurate results. Size distributions of each of The size determination of the nanocapsules and niosomes
these vesicles are exhibited through bar diagrams in Figure 3. was done by dynamic light scattering; the average diameter of
the vesicle was found to be 266.9 nm and 556 nm, respectively.
The distribution patterns are shown in Figure 2.
Trypan Blue Exclusion Test to Study Toxicity of From size determination, the average diameter of micro-
Bacopasaponin-C on Macrophages spheres and liposomes of these vesicles was found to be
Peritoneal macrophages were isolated from thioglycollate 16.28 ¹m and 2.27 ¹m respectively. The size distribution of
injected Balb/c mice in RPMI-1640 medium containing 10% each is demonstrated in Figure 3.
FCS. In each eppendorf containing 500 ¹g of macrophage sus-
pension (5 £ 106 cells), Bacopasaponin-C solution was added
to prepare three effective concentrations (50 ¹g/ml, 250 ¹g, Toxicity of Drug to Macrophages
and 500 ¹g/ml of macrophage suspension). The separate un- The drug was found to be nontoxic to the host peritoneal
treated controls were also prepared. The macrophages were in- macrophages when tested with Trypan blue exclusion test. Re-
cubated with drug for 1 hr at 37± C. Negative control was used sults show negligible macrophage mortality when treated with
with heat-killed macrophages; 500 ¹l of 0.4% Trypan blue so- different drug concentrations, as compared with untreated
lutions was added to the untreated, heat-killed, drug-treated controls (Figure 4).
macrophages and incubated for 10 min. Percent viability for
three sets of treatment and controls were calculated according to
relationship. Assessment of Drug-Induced Leishmania Killing
While testing the drug-induced antileishmanial activity (in
vivo) of the drugs in various forms, free Bacopasaponin-C low-
Cell viability D Total viable cells (unstained)
ered parasitic load of the spleen by 40% whereas liposomal,
£100 =total cells (stained C unstained) niosomal, microencapsulated, and nanocapsulated drug lowered
58 J. SINHA ET AL.

FIG. 3. Size distribution of microspheres (B) and liposomes (A) determined


through light micrometry.

microencapsulated, or nanocapsulated drug. A similar result was


obtained with SGPT level.
Measuring serum urea and creatinine level as shown in
Table 3 assessed nephrotoxicity of the drug. The level of both
urea and creatinine increased in the free drug treatment. But
with the drug in different vesicular forms, values for both urea
and creatinine reduced to normal level, showing apparently no
nephrotoxic effect.

FIG. 2. Size distribution of nanocapsule s (B) and niosomes (A) determined


through dynamic light scattering, using neon laser of ¸ D 632.2 nm, measured
at 90± angle.

the spleen parasite burden by 81%, 86%, 79%, and 91%,


respectively (Table 1).

Assessment of Drug Toxicity


The hepatotoxicity of the drug was explored by assaying
the speciŽ c activities of two liver function enzymes: alkaline
phosphatase (AP) and serum glutamate pyruvate transminase
(SGPT). The results are shown in Table 2. The level of AP
increased on treatment with free drug, but decreased to nor- FIG. 4. Trypan blue exclusion test to study the toxicity of Bacopasaponin- C
mal level when treatment was done with liposomal, niosomal, on macrophages .
BACOPASAPONIN C 59

TABLE 1 TABLE 3
Effect of Bacopasanonin-C loaded liposomes, niosomes, Effect of Bacopasanonin-C loaded liposomes, niosomes,
microspheres, and nanocapsules for treatment of experimental microspheres, and nanocapsules on urea and creatine levels
leishmaniasis on 30-day hamster model related to normal kidney functions

(Parasitic Percent supression Group Ureaa Creatinineb


load in of spleen parasite
Group spleen) £ 10¡9 load Infected untreated control 41.87 § 3.24 0.44 § 0.10
Free drug 50.45 § 2.83 0.58 § 0.15
Infected untreated control 2.65 § 0.24 – Liposomal drug 32.32 § 1.59 0.32 § 0.07
Free drug 1.60 § 0.75 40 Niosomal drug 36.89 § 7.05 0.47 § 0.07
Liposomal drug 0.50 § 0.10 81 Microencapsulated drug 31.97 § 2.61 0.33 § 0.04
Niosomal drug 0.38 § 0.21 86 Nanocapsulated drug 30.68 § 2.88 0.40 § 0.02
Microencapsulated drug 0.54 § 0.07 79
Values are expressed as mean § s.d. (n D 3).
Nanocapsulated drug 0.23 § 0.05 91 a
mg/ml. Normal level: 33.9 § 2.31.
b
Values are expressed as mean § s.d. (n D 3). mg/ml. Normal level: 0.37 § 0.06.

Histopathological Examination
numbers as compared with untreated infected controls.
Histopathological examination (Figure 5) was made through
But in microsphere encapsulated drug treatment (Figure 5D),
light microscopic examination of eosin-haematoxylin stained
both white pulp and red pulp region are of normal disposition. In
sections. Some positive changes were observed for the infected
venous sinuses, number of inŽ ltrating monocytes had reduced.
untreated control (Figure 5A). Comparing with normal ones the
In niosomal drug treatment, both white pulp and red pulp
white pulp region was enlarged and found to be invasive into
regions are of normal structure and the number of inŽ ltrating
red pulp region. Many small bulbs were observed in the venous
monocytes have reduced (Figure 5E).
sinuses. As a whole, the tissue becomes areolar.
In nanocapsulated drug treatment, white pulp region became
In the free drug treatments (Figure 5B), there was regression
much condensed and covered less area. Red pulp is of normal
of the white pulp region, though small white pulp follicles were
disposition. InŽ ltrating manocytes are found in much reduced
still observed. Sinusoidal distribution became nearly normal.
numbers in the venous sinuses (Figure 5F).
InŽ ltration of reticuloendothelial cells or monocytes was still
found. Red pulp region is more or less normal.
In liposomal drug treatment (Figure 5C) the white pulp region
Vesicle Size versus EfŽcacy
became condensed. The follicular artery was distinct, scattered
lymphatic cells were observed, and the red pulp region had nor- The log diameter of each type of vesicle (liposomes, nio-
mal disposition. InŽ ltrating monocytes were found but in less somes, microsphere, and nanocapsules) were plotted against the
parasite-killing efŽ cacy of each corresponding drug-loaded vesi-
cle. We observed that the efŽ cacy was inversely correlated with
TABLE 2
vesicle size (Figure 6). Best efŽ cacy was exhibited with the
Effect of Bacopasanonin-C loaded liposomes, niosomes, smallest vesicle, i.e., the nanocapsules, whereas least efŽ cacy
microspheres, and nanocapsules on speciŽ c enzyme levels was observed with the microspheres. The efŽ cacy gradient was
related to normal liver functions found as:
Serum glutamate
Alkaline pyruvate nanocapsule > niosomes > liposomes > microspheres
Group phosphatase a transaminase b
Infected untreated control 8.71 § 1.84 99.331 § 11.65
Free drug 13.66 § 1.31 121.85 § 13.47
Liposomal drug 8.72 § 0.68 57.02 § 3.25 DISCUSSION
Niosomal drug 9.10 § 0.22 46.3 § 11.60 Saponins have a long history of acting as antimicrobial, an-
Microencapsulated drug 8.89 § 0.45 60.65 § 5.18 tifungal (Otshudi et al. 2000), and antiprotozoal (Traore et al.
Nanocapsulated drug 8.66 § 0.45 61.83 § 5.18 2000; Delmas et al. 2000) agents. Although this is the Ž rst report
Values are expressed as mean § s.d. (n D 3). of Bacopasaponin-C as an antileishmanial agent, the mechanism
a
¹mol p-nitrophenol released/min/dl sera. Normal level: 8.8 § 1.50. of action of Bacopasaponin-C is not known. Besides being an-
b
¹mol of sodium pyruvate released/min L sera. Normal level: 49:6§ tileishmanial in property, it has a terminal glucose moiety and
11:63. as such it is self-targeting in nature to cells having the respective
60 J. SINHA ET AL.

(A) (B)

(C) (D)

(E) (F )

FIG. 5. Histological examination of thin sections of spleen under different experimental conditions (A) infected untreated control, (B) Free drug treated,
(C) liposomal drug treated, (D) microencapsulate d drug treated, (E) niosomal drug treated, and (F) nanocapsulate d drug treated (magniŽ cation £ 400).
BACOPASAPONIN C 61

by encapsulation in appropriate delivery vehicles protects them


from degradation. Encapsulation not only increases their half-
lives, but also increases their efŽ cacy and as such minimizes the
required drug dose.
Besides conventional liposomes and niosomes, polymeric
vesicles have received much attention (Davis and Illum 1988;
Davis et al. 1993; Allerman et al. 1993; Stolnik et al. 1995;
Torchillin and Trubetskoy 1995; Gref et al. 1995). A signiŽ cant
challenge for polymer based drug delivery formulations is the
further development of drug carriers leading to higher therapeu-
tic efŽ cacy. A drug carrier can be efŽ ciently targeted if its size is
below 100 nm diameter (comparable to the size of a virus) and
is not scavenged by the body’s primary defense system such as
FIG. 6. A size versus efŽ cacy relationship of different delivery vesicles: li- the reticuloendothelial system. Obviously, these particles should
posomes, niosomes, microspheres, and nanocapsules . have long circulation times in blood. In addition, the surface of
these nanometer-sized particles should be sufŽ ciently reactive
to chemically bound ligand molecules for receptor-mediated tar-
receptors (Figure 7). On examination, it appears that the leish- geting. The chemical properties of these nanocapsules should be
manicidal property is not due to the inhibitory effect of modulated so that they remain unaffected by circulating lipases,
Bacopasaponin-C on the two vital enzymes of leishmania dono- they should be pH or temperature sensitive, or they should be-
vani viz topoisomerase I and adenosine kinase. The speciŽ c have differently in hydrophilic and hydrophobi c environments.
activity of both the enzymes remains unaltered in the pres- In our research, drug-loaded nanocapsules turned out to be the
ence of varying concentrations of Bacopasaponin-C (Ray et al. most efŽ cacious in killing parasites; the least effective was the
1998; Dutta et al. 1987). Moreover, its leishmanidal property drug-loaded microspheres, the largest vesicular delivery system
is parasite-speciŽ c as judged from Trypan blue exclusion test tried so far. An inverse linear relationship between the size of
involving both the host cells and the parasites. the vesicles and percent efŽ cacy in killing the parasites was
A wide range of bioactive molecules are commercially avail- established suggesting that composition and the size are the two
able as drugs. Many of these are stable only under physiological important parameters that must be considered for a strategy for
conditions and their therapeutic application is severely limited efŽ cient delivery.
by their short half-lives in vivo. Most of the administered amount
does not reach the target site. Thus, delivery of these molecules
REFERENCES
Allerman, E., Gurny R., and Doelker, E. 1993. Drug loaded nanoparticules :
Preparation, methods and drug targeting tissues. Eur. J. Pharm. Biopharm.
39:173 – 191.
Anderson, J. M., and Shive, M. S. 1997. Biodegradation and biocompatibility
of PLA and PLGA microsphere. Adv. Drug. Del. Rev. 28:5 – 24.
Baillie, A. J., Florence, A. T., Hume, L., Muirhead G. T., and Rogerson, A. 1985.
The preparation and properties of niosomes-non-ioni c surfactant vesicles.
J. Pharm. Pharmacol. 37:863 – 868.
Davis, S. S., and Illum, L. 1988. Polymeric microspheres as drug carriers.
Biomaterials 9:111 – 115.
Davis, S. S., Illum, L., Mognimi, S. M., Davies, M. C., Porter, C. J. H., Muir,
I. S., Brindley, A., Christy, N. M., Norman, M. E., Williums, P., and Dunn,
S. E. 1993. Microspheres for targeting drugs to speciŽ c body sites. J. Cont.
Release 24:157 – 163.
Delmas, F., Giorgio, C., Elias, D. R., Gasquet, M., Azas, N., Mshvildadze, V.,
Dekanosidze , G., Kemertelidze, E., and David, P. T. 2000. Antileishmanial
activity of three saponins isolated from ivy, alpha hederin, beta hederin and
hederacol chiside AI, as compared to their action on mammalian cells cultured
in vitro. Planta Medica 66(4):343 – 347.
Dutta, A. K., Bhaumik, D., and Chatterjee, R. 1987. Isolation and characteriza-
tion of Adenosine Kinase from Leishmania donovani . J. Biol. Chem. 262 (12):
5515 – 5521.
Garai, S., Mahato, S. B., Ohtani, K., and Yamasaki, K. 1996. Damnarane type
triterpenoid saponins from Bacopa monniera. Phytochemistry 42(3):815 – 820.
Gref, R., Domb, A., Quelle, P., Blunk, T., Muller, R. H., Verbatz, J. M., and
Langer, R. 1995. Controlled intervenous delivery of drugs using PEG-coated
FIG. 7. Structure of Bacopasaponin-C . sterically stabilized nanospheres . Adv. Drug Del. Rev. 16:215 – 233.
62 J. SINHA ET AL.

Gregoriadis, G., and Ryman, B. E. 1972. Lysosomal, localization of fracto Ray, S., Hazra, B., Mittra, B., Das, A., Mazumder, H. K., and Diospyrin, A.
furanosidase containing liposomes injected into rats. Biochem. J. 129: 1998. Bisnaphthoquinone : A novel inhibitor of Type I DNA topoisomerase
123 – 137. leishmania donovani . Mol. Pharmacol. 54:994 – 999.
Guerrero, D. Q., Allemann, E., Fessi, H., and Doelker, E. 1998. Preparation Tech- Reitman, S., and Frenkel, S. A. 1957. Colorimetric method for the determination
niques and mechanism of formation of Biodegradabl e Nanoparticles from of serum glutamic oxalacetic acid and glutamic pyruvic transaminase. Am. J.
preformed polymers. Drug Dev. Ind. Pharmacy 24(12):1113 – 1128. Clin. Path. 28:53 – 56.
Gurr, E. 1962. Staining, animal tissues practical and theoretical. London: Seyler, I., Morin, C., Barratt, G. Appel, M., Devissaguet, J. P., and Puisieux,
Leonard Hill. F. 1995. Induction of macrophag e NO-synthase by an immunomodulato r en-
Guterres, S. S., Fessi, H., Barratt, G., Devissaguet J. P., and Puisieux, F. 1995. trapped within polymeric nanocapsules . Eur. J. Pharm. Biopharm. 41(1):49–
Poly (DL-lactide) nanocapsule s containing diclofenac: I. Formulation and 54.
stability study. Int. J. Pharm. 113:57 – 63. Stauber, L. A., Franchino, E., and Grun, J. 1958. An eight-day method for screen-
Hiremath, S. R., and Hota, A. 1999. Nanoparticles as drug delivery systems. ing compoun d against Leishmania donovan i in golden hamster. J. Protozool.
Indian J. Pharm. 61(2):69 – 75. 5:263 – 273.
Kind, P. R. N., and King, E. J. 1954. Estimation of plasma phosphatas e by Stolnik, S., Illum, L., and Davis, S. S. 1995. Long circulating microparticulate
determination of hydrolysed phenol with amino antipyrine. J. Clin. Path. 7: drug carrier. Adv. Drug Del. Rev. 16:195 – 214.
322 – 326. Torchilin, V. P., and Trubetskoy, V. S. 1995. Which polymers can make nanopar-
Kreuter, J. 1996. Nanoparticles and microparticles for drug and vaccine delivery. ticulate drug carriers long circulating. Adv. Drug Del. Rev. 16:141 – 145.
J. Anat. 189(Pt.3):503 – 505. Traore, F., Faure, R., Olliver, E., Gasquet, M., Azas, N., Debrauwer, L., Keita,
Majhi, P. R., and Moulik, S. P. 1999. Microcalorimetric investigations of AOT A., David, P. T., and Balansard, G. 2000. Structure and antiprotozoal activity
self-association in oil and state of pool water in water/oil microemulsions . of triterpenoid saponins from Glinus oppositifolius. Planta Medica 66(44):
J. Phys. Chem. 103:5977 – 5983. 368 – 371.
Moulik, S. P., De, G. C., Panda, A. K., Bhowmick B. B., and. Das, A. R. 1999. Wang, Min Ya., Sato, H., Adachi I., and Horikoshi, I. 1996. Preparation and
Disperse molecular aggregates, 1. Synthesis and characterization of nanopar- characterization of poly (lactic-co-glycoli c acid) microspheres for targeted
ticles of Cu2 [Fe(CN)6 ] in H2 O/AOT/W- heptane water-in-oil microemulsion delivery of novel anticancer agent. Taxol. Chem. Pharm. Bull. 44(10):1935 –
media. Langmuir 15:8361 – 8367. 1940.
Otshudi, A. L., Foriers, A., Vercruysse, A., Zeebroeck, A. V., and Lauwers, S. Yoshioka, T., Sternberg B., and Florence, A. T. 1994. Preparation and properties
2000. In vitro antimicrobiol treatment of dysentery and diarrhoea in Demo- of vesicles (niosomes ) of sorbitan monoesters (span 20, 40, 60 and 80) and
cratic Republic of Congo (DRC). Phytomedicine 7(2):167 – 172. sorbitan triesters (span 85). Int. J. Pharm. 105:1 – 6.

You might also like