Antigen Presenting Cells Targeting and Stimulation Potential of

Feb 11, 2015 - AmB was kindly provided as a gift sample from Emcure pharmaceutical Ltd. (Pune, India). All other reagents were of analytical grade. Pa...
5 downloads 0 Views 5MB Size
Subscriber access provided by UNIVERSITY OF MICHIGAN LIBRARY

Article

Antigen presenting cells targeting and stimulation potential of lipoteichoic acid functionalized lipo-polymerosome: A chemoimmunotherapeutic approach against intracellular infectious disease Pramod Kumar Gupta, Anil K. Jaiswal, Shalini Asthana, Anuradha Dube, and Prabhat Ranjan Mishra Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/bm5015156 • Publication Date (Web): 11 Feb 2015 Downloaded from http://pubs.acs.org on February 18, 2015

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Biomacromolecules is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

100x51mm (300 x 300 DPI)

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 43

1 2 3

Antigen presenting cells targeting and stimulation potential of lipoteichoic acid functionalized lipo-polymerosome: A chemo-immunotherapeutic approach against intracellular infectious disease

4

Pramod K. Guptaa, Anil K. Jaiswalb, Shalini Asthanaa, Anuradha Dubeb and Prabhat R. Mishraa*

5 6

a

7 8

b

9 10

Pharmaceutics Division, Council of Scientific and Industrial Research-Central Drug Research Institute, B 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, India 226031 Parasitology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, B 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, India 226031 Email: [email protected], [email protected], [email protected], [email protected], [email protected]

11 12 13 14 15 16 17 18 19

* Corresponding Author

20 21 22 23 24 25 26 27 28 29 30 31

Prabhat R. Mishraa Principal Scientist Pharmaceutics Division, Preclinical South PCS 002/011, CSIR-Central Drug Research Institute B 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow-226031, India Phone :91-522-2771940; 2771942 Fax: 91-522-2771941

32 1

ACS Paragon Plus Environment

Page 3 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

33

Abstract

34

Antigen presenting cells (APC) are well-recognized therapeutic targets for intracellular

35

infectious diseases including visceral leishmaniasis. These targets have raised concerns regarding

36

their potential for drug delivery due to over expression of a variety of receptors for pathogen

37

associated molecular pathways after infection. Since, Lipoteichoic acid (LTA), a surface

38

glycolipid of gram-positive bacteria responsible for recognition of bacteria by APC receptors

39

which also regulate their activation for pro-inflammatory cytokine secretion, provides additive

40

and significant protection against parasite. Here, we report the nanoarchitechture of APC focused

41

LTA functionalized Amphotericin B encapsulated lipo-polymerosome (LTA-AmB-L-Psome)

42

delivery system mediated by self assembly of synthesized glycol chitosan-stearic acid copolymer

43

(GC-SA) and cholesterol lipid, which can activate and target the chemotherapeutic agents to

44

Leishmania parasite resident APC. Greater J774A and RAW264.7 macrophage internalization of

45

FITC tagged LTA-AmB-L-Psome compared to core AmB-L-Psome was observed by

46

FACSCalibur cytometer assessment. This was further confirmed by higher accumulation in

47

macrophage rich liver, lung and spleen during biodistribution study. The LTA-AmB-L-Psome

48

overcame encapsulated drug toxicity and significantly increased parasite growth inhibition

49

beyond commercial AmB treatment in both in vitro (macrophage-amastigote system; IC50,

50

0.082±0.009 µg/mL) and in vivo (Leishmania donovani-infected hamsters; 89.25±6.44% parasite

51

inhibition) models. Moreover, LTA-AmB-L-Psome stimulated the production of protective

52

cytokines like interferon-γ (IFN-γ), interleukin-12 (IL-12), tumor necrosis factor-α (TNF-α) and,

53

inducible nitric oxide synthase and nitric oxide with down-regulation of disease susceptible

54

cytokines like transforming growth factor-β (TGF-β), IL-10 and IL-4. These data demonstrate

55

the potential use of LTA functionalized lipo-polymerosome as a biocompatible lucrative 2

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 43

56

nanotherapeutic platform for overcoming toxicity and improving drug efficacy along with

57

induction of robust APC immune responses for effective therapeutics of intracellular diseases.

58 59

Keywords: Leishmania, amastigotes, cytokines, lipo-polymerosome, biodistribution, toxicity

60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 3

ACS Paragon Plus Environment

Page 5 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

81

1. Introduction

82

The professional antigen presenting cells (APC), i.e. monocytes/macrophages, B-cells and

83

dendritic cells are major port of survival of parasites into the body and plays an important role in

84

disease progression 1. The intracellular parasite utilizes immune evasion mechanisms by down-

85

regulation of parasite antigen presentation via the MHC class II antigen pathway and prevents

86

activation of oxidative bursts and other microbicidal mechanisms

87

eruption of many diseases including tuberculosis, leishmaniasis, cryptococcosis and filariasis.

88

Despite numerous advances over the past decades, effective intra-APC disease chemotherapy

89

remains challenging. The generalized toxicity of many chemotherapeutic drugs makes it difficult

90

to achieve therapeutic concentrations without systemic side effects. Consequently, architecture of

91

such drug delivery system is essential that preferentially recognize and bind to specific over

92

expressed receptors on infected APC surface, with the hope that the delivery system will be

93

internalized and release its contents specifically in to the cell 4. Such drug vehicles could

94

potentially open the door to new therapeutic paradigms for practicing medicine that promises to

95

eradicate a range of parasitic diseases. Additionally, the toxicity related issues of drug can be

96

reduced by targeted delivery to intracellular regions of APC.

97

Taking together the immuno-stimulatory tendency of bacteria and its forced engulfment by APC,

98

bacterial antigen can be utilized as a component of drug delivery system that will have artificial

99

bacteria like identity. Importantly, such delivery system suites as a platform for biomarker

100

specific ligand display and affinity selection, raising the possibility that a single vehicle can be

101

used both for identification of cell-targeting and immunostimulatory ligand for specific delivery

102

of drug.

2, 3

. These events lead to

4

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 43

103

APC expresses a variety of receptors for pathogen associated molecular pathways (PAMPs) such

104

as lipopolysaccharides, glycoproteins and a range of polyanionic molecules, which get over

105

expressed after infection. These enable the recognition and phagocytosis of large repertoire of

106

pathogenic parasites and subsequent generation of inflammation to eradicate the infection.

107

Since, Lipoteichoic acid (LTA) is a common immunostimulatory polyanionic cell-wall

108

component of Staphylococci and many gram-positive bacteria, composed of poly(glycerol

109

phosphate) attached to the glycolipid anchor β-D -GlcpII-(1→6)-β-D-GlcpI-(1→3)-diacylglycerol

110

5, 6

111

create a negatively charged network which extends from the membrane to the surface of the

112

bacterium and is recognized by involvement of scavenger receptor (SR) particularly type 1 7,

113

together with Toll-like receptors (TLRs) especially TLR2 and TLR6, and its co-receptors (CD14,

114

CD36), of APC which results in induction of cytokines and co-stimulatory molecules for antigen

115

presentation that can modulate T cell polarization. TLRs specific ligands have ability to enhance

116

drug efficacy through multiple mechanisms: TLRs stimulation of APC results in increase in

117

surface peptide/MHC complexes, co-stimulatory molecules, and cytokine secretion, the three

118

signals required for T cell activation and proliferation 8. Additionally, LTA bound to APC can

119

interact with circulating antibodies and activate the complement cascade to induce passive

120

immune kill phenomenon 9. The rationale for developing this prototype formulation i.e.

121

Lipoteichoic acid modified lipopolymerosome (LTA-AmB-L-Psome) is to target the carrier

122

system to Antigen Presenting Cells (APCs) which are actually reservoir of Leishmania parasites

123

and finally to impart immunostimulatory potential (being component of bacterial cell wall) to

124

carrier system so as to evoke Th1 mediated killing of Leishmania parasites. This dual strategy is

. Because of their intrachain phosphodiester groups, LTA collectively with teichoic acids

5

ACS Paragon Plus Environment

Page 7 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

125

likely to improve pharmacokinetics and reduce toxicity to a greater extent by preventing AmB

126

exposure to other organs.

127

Previously, we have reported Lipo-polymerosome (L-Psome) by spontaneous self-assembly of

128

synthesized glycol chitosan-stearic acid copolymer as stable Amphotericin B (AmB) delivery

129

system able to reduce toxicity issues 10. In the present study, we have demonstrated better AmB

130

chemotherapy for leishmaniasis through immuno-modulation using LTA as specific ligand

131

anchored on lipo-polymerosome (LTA-AmB-L-Psome). Furthermore, the developed formulation

132

has potential for its application in the condition of VL and intra-APC diseases.

133

2. Material and methods

134

2.1. Materials

135

Glycol

136

(dimethylamino)propyl]carbodiimide hydrochloride (EDC), Dimethyl acetamide (DMAc)

137

sodium deoxycholate (SDC), stearic acid (SA), cholesterol, fluorescein isothiocynate (FITC),

138

Dulbecco’s modified Eagle’s medium (DMEM), Roswell Park Memorial Institute (RPMI) 1640

139

medium, streptomycin, fetal bovine serum (FBS) and LTA from Bacillus subtilis were purchased

140

from Sigma-Aldrich (St. Louis, MO). Water used in all the experiments was obtained by Milli-Q

141

Plus 185 water purification system. Methanol and acetonitrile of HPLC grade were provided by

142

SD Fine Chem Ltd (Mumbai, India). AmB was kindly provided as a gift sample from Emcure

143

pharmaceutical Ltd. (Pune, India). All other reagents were of analytical grade.

144

Parasite and cell line culture

145

The WHO reference strain of L. donovani (MHOM/IN/80/Dd8) was used for both in vitro and in

146

vivo experiments. Leishmania parasites and the macrophage cell line J774A, RAW264.7 were

147

maintained in RPMI-1640 medium (Sigma, USA) supplemented with 10% heat inactivated fetal

chitosan

(GC,

Mw

90

KD,

82.7%

degree

of

deacetylation),

[1-ethyl-3

6

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 43

148

bovine serum (HIFBS), 100 U/ml penicillin and 100 µg/ml streptomycin at 37 oC in humidified

149

atmosphere of 5% (v/v) CO2/air mixture.

150

Animals and ethics statements

151

For in vivo antileishmanial study, Syrian golden male hamsters (Mesocricetus auratus, 45-50 g),

152

for pharmacokinetic and bio-distribution study, male Wistar rats (180-200 g) animal models

153

were used while biochemical analysis and tissue histopathology performed in male Swiss mice

154

(18-20 g) with prior approval of the Animal Ethics Committee of CSIR-Central Drug Research

155

Institute and according to regulations of the Council for the Purpose of Control and Supervision

156

of Experiments on Animals (CPCSEA), Ministry of Social Justice and Empowerment, Govt. of

157

India. The Institutional Animal Ethics Committee (IAEC) approval no. is CDRI/2012/38.

158

2.2. Preparation of lipoteichoic acid modified lipo-polymerosome (LTA-AmB-L-Psome)

159

The core self assembled L-Psome formulation was prepared through a nano-precipitation method

160

followed by sonication as illustrated in our previous work

161

synthesized glycol chitosan-stearate copolymer through carbodiimide coupling reaction between

162

glycol chitosan and stearic acid in molar ratio of 4:1.

163

Thereafter, synthesized copolymer (4 mg), AmB (1 mg in 100 µL of DMAc), and cholesterol (1

164

mg) was taken in 250 µL of ethanol and were mixed gently. This solution added drop wise in 1

165

mL of distilled water under constant stirring for 5 min in 5 mL vial. Obtained dispersion was

166

sonicated using probe sonicator (misonix, Germany) at 20% amplitude for 120 sec (15 sec pulse

167

on and 5 sec pulse off), followed by dialysis against 1000 mL water for 2 h for the purpose to

168

remove un-entrapped AmB and solvents.

169

In order to prepare LTA-AmB-L-Psome, the exterior surface of the L-Psome was modified using

170

LTA ligand by adsorption method. For this purpose, 5 mg L-Psome was incubated with 1 mL

10

. For this purpose, firstly, we

7

ACS Paragon Plus Environment

Page 9 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

171

LTA solution (at different concentrations from 0.02 to 0.4% w/v) in 10 mM HEPES buffer (pH

172

7.4) at 37 oC for 24 h to allow LTA to become electrostatically anchor onto L-Psome surface.

173

The excess LTA was removed by washing followed by centrifugation of the resultant

174

suspension, and the whole process was repeated thrice to ensure complete removal. The LTA

175

amount was optimized by analyzing zeta potential change in LTA-L-Psome dispersion at 25 oC

176

as shown in Fig. 1(a).

177

2.3. Physicochemical Characterization of L-Psome

178

The mean particle size, size distribution and zeta potential of the L-Psome were determined with

179

a Zetasizer Nano ZS (Malvern Instruments, Worcestershire, UK) at 25 oC after dilution in

180

distilled water.

181

The surface morphology of optimized LTA-L-Psome was ascertained using high resolution

182

transmission electron microscopy (HR-TEM, Tecnai™ G2 F20, Eindhoven, The Netharlands) 10.

183

To quantify LTA anchoring to L-Psome, BCA method for direct estimation of protein was

184

performed. In this method, initially LTA decorated L-Psome eluted from sephadax G100 silica

185

Gel column then measured by NANODROP 2000 spectrophotometer (Thermo Scientific, USA)

186

11-14

187

% Conjugation efficiency = Conjugated wt. of LTA × 100 / Total wt. of L-Psome

188

The solvent injection method was utilized to load the AmB within LTA-L-Psome, for this AmB

189

solution in DMAc was added in LTA-L-Psome with constant stirring. The AmB amount

190

encapsulated in LTA-L-Psome was quantified as the amount of un-entrapped drug recovered in

191

the supernatant by reverse phase HPLC

192

subsequent washings of the formulation. The percentage of drug loading, encapsulation

193

efficiency (EE) was calculated using following formulae:

. The coupling efficiency was expressed as the percentage of LTA bound to L-Psome surface

15

after ultracentrifugation at 40,000×g for 30 min and

8

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 43

194

Loading efficiency = Wt. of (Total – free) AmB in L-Psome × 100/ Wt. of L-Psome

195

Encapsulation efficiency = Wt. of (Total – free) AmB in L-Psome × 100/ Wt. of Total AmB

196

The release of AmB from formulations was assessed using a dialysis membrane under sink

197

conditions. Herein, 2 mg AmB equivalent formulations (LTA-AmB-L-Psome AmB-L-Psome,

198

Fungizone, and Ambisome) was kept in dialysis bag, sealed and placed in 200 ml of release

199

medium with 2% w/v SDC, under continuous shaking at 100 rpm at 37 °C. A definite volume of

200

the release medium was withdrawn at regular time intervals (30 min and 1, 2, 3, 4, 6, 8, 12, 24,

201

48, 72 h) and replaced with equal volume of fresh medium. Amount of the AmB in the collected

202

samples was analyzed using described HPLC method.

203

To understand stability of LTA-AmB-L-Psome, stability study was carried out at different time

204

points (1, 7, 15, 30, 90 and 180 days) in PBS at 2-8 0C, 37 0C and room temperature along with

205

other formulations i.e. AmB-L-Psome, Ambisome and Fungizone. At each time point, an aliquot

206

of each formulation was withdrawn for analysis to observe any change in physicochemical

207

parameters such as size and EE of the formulation 10.

208

Furthermore, rigid characteristic of LTA-AmB-L-Psome was evaluated through HRTEM study.

209

We have observed change in surface morphology of vesicles after storage period of 6 months

210

with 10 % w/v BSA and 10 % v/v Wistar rat plasma at 2-8 0C. In continuation to this, rigidity of

211

LTA-AmB-L-Psome was also evaluated by analyzing size and PDI variation, when incubated

212

with 10 % w/v BSA and 10 % v/v rat plasma at RT under gentle stirring at a concentration of 1

213

mg AmB/mL. At predetermined intervals up to 60 min sample was taken out and measurements

214

were performed in triplicate.

215

2.4. Tagging of AmB with FITC 9

ACS Paragon Plus Environment

Page 11 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

11

216

For in vitro uptake studies, AmB was tagged with FITC as previously reported method

.

217

Briefly, AmB (10 mg) and FITC (5 mg) were dissolved in DMAc (2 mL) followed by addition of

218

200 µL triethylamine as base catalyst in 5 mL round bottom flask, followed by 2 h stirring at

219

room temperature and thereafter 10 mL ethyl acetate was added to precipitate the compound.

220

Colloidal precipitate was separated by centrifugation (18,000×g for 10 min) and dried over

221

desiccant under vacuum. In order to verify tagging of FITC with AmB (FAmB) TLC analysis

222

was carried out using a mobile phase composed of ethyl acetate: methanol (2:3).

223

2.5. In vitro uptake by macrophages

224

FAmB was encapsulated in the LTA-L-Psome using same procedure as described earlier, in

225

short, 1 mg of tagged FAmB was dissolved in 100 µL of DMAc and loaded drop wise in blank

226

LTA-L-Psome. Infected J774A and RAW264.7 macrophage cells in 2×105 cells/well were

227

placed separately on to 12-well cluster dish and cultivated in 800 µL DMEM supplemented with

228

10% FBS and 1% antibiotic and antimycotic. After 24 h the culture medium was replaced with

229

fresh culture medium. Cells were incubated for 6 h at 37 oC and 5% CO2 with FITC tagged

230

formulations (LTA-FAmB-L-Psome and FAmB-L-Psome) at 10 µg/mL concentration. After

231

incubation, cells were transferred in vials and relative fluorescence was measured by

232

FACSCalibur (Becton Dickinson, Oxford, UK) at λEX (495 nm) and λEM (525 nm). FACSCalibur

233

mediated macrophage uptake study was also carried out for non-infected as well as LTA

234

saturated infected J774A and RAW264.7 macrophage cells.

235

Macrophage uptake was also assessed by confocal laser scanning microscopy (CLSM). J774A

236

cells seeded at a density of 1×105 cells/well on poly-L-lysine-coated glass cover slips (in six-well

237

plates), were left for 12 h and then incubated for 4 h with LTA-FAmB-L-Psome and FAmB-L-

238

Psome at room temperature in the dark followed by repeated washing and fixed in 10% formalin 10

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 43

239

in PBS and observed by CLSM (Carl Zeiss LSM5100, Germany) equipped with ×40 objective

240

lens. λEX (495 nm) and λEM (525 nm) of FITC were used to analyze tagged AmB.

241

2.6. Pharmacokinetic and Bio-distribution study

242

Wistar rats (180-200 g) were allocated into the following four treatment groups: IV bolus

243

administration of 5 mg/kg AmB-L-Psome (n=6), 5 mg/kg LTA-AmB-L-Psome (n=6), 1 mg/kg

244

Fungizone (n=6) and 5 mg/kg AmBisome (n=6). The doses selected for IV administration of

245

commercial AmB formulations in present study are in the range for which there is no clinical

246

nephrotoxicity for each formulation, based on published reports

247

was sampled at 10 min pre-dose and 0.5, 1, 2, 3, 5, 8, 12, 24, 48 and 72 h post-dose of IV bolus

248

administrations of AmB formulations, followed by separation of plasma by centrifugation at

249

2,500×g for 10 min at 15 oC). The animals were sacrificed at 0.5, 1, 2, 3, 5, 8, 12, 24, 48 and 72

250

h post administration of AmB formulations, and liver, spleen, right lung, right kidney and heart

251

were harvested for drug analysis. Plasma and tissue samples were stored at -80 oC until drug

252

analysis.

16

. Systemic blood (0.25 mL)

253

Samples were analyzed using Shimadzu HPLC system equipped with 10 ATVP binary

254

gradient pumps (Shimadzu, Japan), a Rheodyne model 7125 injector (CA, US) with a 20 µL loop

255

and SPD-M10 AVP UV detector (Shimadzu, Japan). AmB in plasma and tissue samples was

256

analyzed following published validated isocratic HPLC method with slight modifications,15

257

using column [LichroCART® 250-4, Lichrospher® 100, RP-18e, 5 µm, 250×4 mm (Merck

258

KgaA, 64271 Darmstadt, Germany)] and mobile phase [acetonitrile/10mM KH2PO4 buffer, pH 4

259

(60:40, v/v)] at a flow rate of 1 mL/min. Tissue samples were homogenized in acetonitrile (1 g

260

tissue per 2 mL acetonitrile) using IKA T25 digital ULTRA-TURRAX for 3-4 min over an ice

261

bath. The filtered aliquots of 20 µL of processed plasma and tissues were used for quantitative 11

ACS Paragon Plus Environment

Page 13 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

262

analysis. Calibration curves of AmB were linear in the range of 0-10 µg/mL for plasma and 0-10

263

µg/g for tissue samples. The AmB quantification limit for plasma was 20 ng/mL and for tissue

264

samples was 20 ng/g.

265

2.7. In vitro anti-amastigote activity

266

The activity of AmB-formulations against intracellular amastigotes was evaluated as per protocol

267

described earlier

268

infected with green fluorescent protein (GFP) expressed promastigotes at multiplicity of 10

269

parasites per macrophage. After 12 h incubation 24-well plates were washed thrice with PBS (pH

270

7.4) to remove non-phagocytosed promastigotes and re-supplemented with RPMI-1640 complete

271

medium, followed by incubation with LTA-AmB-L-Psome, AmB-L-Psome, Ambisome and

272

Fungizone at different drug concentrations, and with the same amount of blank formulations in

273

triplicate. After 48 h incubation, cells were removed, washed in PBS and quantitated by

274

FACSCalibur cytometer equipped with a 20 mW argon laser at 488 nm excitation and 515 nm

275

emission followed by data analysis using Kaluza analysis software (Beckman Coulter). The

276

parasite growth inhibition was determined by relative fluorescence levels of drug-treated

277

parasites with that of untreated control parasites and the inhibitory concentrations (IC50 and IC90)

278

were calculated using GraphPad Prism6.

279

2.8. In vivo assay in L. donovani infected hamsters

280

The in vivo AmB-formulation efficacy was studied against L. donovani amastigotes in a golden

281

hamster model

282

intraperitoneally dosed 1 mg/kg of LTA-AmB-L-Psome, AmB-L-Psome, Fungizone and

283

Ambisome each for 5 consecutive days. Two week post treatment, treated animals were

284

sacrificed and results were compared with the infected untreated control group. Detection of

10

17

. Briefly, J774A macrophages (1×105 cells/well) in 24-well plates were

. After 30 days of established infection, hamsters (n = 5 in each group) were

12

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 43

285

parasitic load in spleen was carried out by blinded microscopic enumeration with Giemsa-stained

286

spleen impression smear. Parasite burdens were calculated by counting the number of

287

amastigotes per 100 macrophage nuclei. The percentage of inhibition (PI) was calculated using

288

the formula:

289

PI= (PP-PT/PP) ×100

290

Where PP and PT are number of amastigotes per 100 macrophage nuclei before and after

291

treatment, respectively.

292

2.9. Nitric oxide (NO) determination

293

NO content in the splenocytes culture supernatants of treated hamsters was monitored by Griess

294

assay method

295

sulfanilamide 1N HCl and 0.1% N-(1-naphthyl) ethylenediamine dihydrochloride in H2O] were

296

mixed at 1:1 ratio and incubated for 15 min at room temperature, and the absorbance was

297

determined at 540 nm by microplate reader (DTX 800 multimode detector, Beckman Coulter).

298

Nitrite concentration was calculated with a sodium nitrite standard curve generated for each

299

experiment.

300

2.10. Evaluation of Th1/Th2 cytokines and chemokine

301

Splenic cell smear of treated and untreated control groups were analyzed for various cytokines

302

(TNF-α, IL-12, IFN-γ, IL-4, IL-10 and TGF-β) and inducible nitric oxide synthase (iNOS) using

303

quantitative real time-PCR (qRT-PCR) technique. Splenocytes mRNA of different hamster

304

groups was isolated using Tri reagent (Invitrogen, USA) followed by cDNA synthesis using first

305

strand cDNA synthesis kit (Fermentas, USA) as per manufacturer’s protocol. qRT-PCR was

306

conducted with initial denaturation at 95 °C for 2 min followed by 40 cycles, each consisting of

307

denaturation at 95 °C for 30s, annealing at 55 °C for 40 s, and extension at 72 °C for 40 s per

18

. Briefly, the culture supernatant and the mixture of Greiss reagent [1%

13

ACS Paragon Plus Environment

Page 15 of 43

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

308

cycle using the iQ5 multicolor real-time PCR system (Bio-Rad, USA). Comparator samples were

309

the cDNAs from infected hamsters. The PCR signals quantification was performed by comparing

310

the cycle threshold (CT) value of the gene of interest with that of reference gene hypoxanthine

311

phosphoribosyltransferase (HPRT). Results were expressed as fold change (FC) of mRNA

312

relative to those in un-stimulated cells.

313

2.11. Toxicity assay

314

The hemolysis and J774A cell cytotoxicity assay was performed in 5 to 20 µg/ml AmB

315

concentrations of LTA-AmB-L-Psome, AmB-L-Psome and their equivalent blank formulations

316

using methods reported previously 15. Subacute toxicity assay was performed in five mice groups

317

(n=3), received LTA-AmB-L-Psome (5mg/kg), AmB-L-Psome (5mg/kg), Ambisome (5mg/kg),

318

Fungizone (1mg/kg) and saline (control group) intravenously in a constant volume of 200 µL

319

daily for 15 days. Twelve hours post treatment, animals were euthanized, and blood was

320

collected by cardiac puncture, centrifuged (2000 × g) and subsequently analyzed for biochemical

321

parameters. Transaminase activities, urea and creatinine were determined using an automatic

322

Hitachi 912 apparatus (Roche Diagnostics Corporation, Indianapolis, IN, USA). Kidney, liver,

323

spleen, lung, heart and colon tissues were collected for histopathological studies as reported in

324

our previous publication 10.

325

2.12. Statistical analysis

326

All results are presented as mean±standard deviation (S.D.) of three independent measurements.

327

Data were analyzed by one-way analysis of variance (ANOVA) and p value of